WO2015118175A2 - Inhibition ciblee de facteur de croissance de transformation bêta (tgfβ) - Google Patents

Inhibition ciblee de facteur de croissance de transformation bêta (tgfβ) Download PDF

Info

Publication number
WO2015118175A2
WO2015118175A2 PCT/EP2015/052781 EP2015052781W WO2015118175A2 WO 2015118175 A2 WO2015118175 A2 WO 2015118175A2 EP 2015052781 W EP2015052781 W EP 2015052781W WO 2015118175 A2 WO2015118175 A2 WO 2015118175A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
tgfp
tumor
group
protein
Prior art date
Application number
PCT/EP2015/052781
Other languages
English (en)
Other versions
WO2015118175A3 (fr
Inventor
Kin-Ming Lo
Original Assignee
Merck Patent Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2015213988A priority Critical patent/AU2015213988B2/en
Priority to SG11201606577YA priority patent/SG11201606577YA/en
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Priority to NZ721364A priority patent/NZ721364A/en
Priority to EP21165709.3A priority patent/EP3889172A1/fr
Priority to EP15717097.8A priority patent/EP3105246B1/fr
Priority to DK15717097.8T priority patent/DK3105246T3/da
Priority to RS20210783A priority patent/RS62038B1/sr
Priority to SI201531639T priority patent/SI3105246T1/sl
Priority to KR1020167024679A priority patent/KR102363008B1/ko
Priority to CA2934979A priority patent/CA2934979A1/fr
Priority to PL15717097T priority patent/PL3105246T3/pl
Priority to ES15717097T priority patent/ES2876430T3/es
Priority to CN201580007865.3A priority patent/CN106103488B/zh
Priority to BR112016014952A priority patent/BR112016014952A2/pt
Priority to PE2020002093A priority patent/PE20210168A1/es
Priority to LTEP15717097.8T priority patent/LT3105246T/lt
Priority to RU2016135062A priority patent/RU2752424C2/ru
Priority to JP2016546949A priority patent/JP6731346B2/ja
Priority to MX2016010067A priority patent/MX2016010067A/es
Publication of WO2015118175A2 publication Critical patent/WO2015118175A2/fr
Publication of WO2015118175A3 publication Critical patent/WO2015118175A3/fr
Priority to IL246968A priority patent/IL246968B/en
Priority to PH12016501549A priority patent/PH12016501549A1/en
Priority to AU2019246876A priority patent/AU2019246876B2/en
Priority to IL281565A priority patent/IL281565B/en
Priority to HRP20210977TT priority patent/HRP20210977T1/hr
Priority to CY20211100564T priority patent/CY1124408T1/el
Priority to AU2022200365A priority patent/AU2022200365A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • This invention relates generally to bifunctional molecules including (a) a TGFpRII or fragment thereof capable of binding TGFP and (b) an antibody, or antigen binding fragment thereof, that binds to an immune checkpoint protein, such as Programmed Death Ligand 1 (PD- Ll), uses of such molecules (e.g., for treating cancer), and methods of making such molecules.
  • a TGFpRII or fragment thereof capable of binding TGFP and (b) an antibody, or antigen binding fragment thereof, that binds to an immune checkpoint protein, such as Programmed Death Ligand 1 (PD- Ll)
  • PD- Ll Programmed Death Ligand 1
  • cancer immunotherapy is a new paradigm in cancer treatment that instead of targeting cancer cells, focuses on the activation of the immune system. Its principle is to rearm the host's immune response, especially the adaptive T cell response, to provide immune surveillance to kill the cancer cells, in particular, the minimal residual disease that has escaped other forms of treatment, hence achieving long-lasting protective immunity.
  • the present invention is based on the discovery that a bifunctional protein containing at least portion of TGFp Receptor II (TGFpRII) that is capable of binding TGFp and antibody or antigen-binding fragment that binds to an immune checkpoint protein such as human protein Programmed Death Ligand 1 (PD-L1) can be an effective anti-tumor and anti-cancer therapeutic.
  • TGFpRII TGFp Receptor II
  • PD-L1 human protein Programmed Death Ligand 1
  • the protein can exhibit a synergistic effect in cancer treatment, as compared to the effect of administering the two agents separately.
  • the present invention features a protein including (a) human TGFpRII, or a fragment thereof capable of binding TGF (e.g., a soluble fragment); and (b) an antibody, or an antigen-binding fragment thereof, that binds PD-L1 (e.g., any of the antibodies or antibody fragments described herein).
  • the invention features a polypeptide including (a) at least a variable domain of a heavy chain of an antibody that binds PD-L1 (e.g., amino acids 1-120 of SEQ ID NO: 2); and (b) human TGFPRII, or a soluble fragment thereof capable of binding TGFp (e.g., a human TGFpRII extra-cellular domain (ECD), amino acids 24-159 of SEQ ID NO: 9, or any of those described herein).
  • the polypeptide may further include an amino acid linker connecting the C-terminus of the variable domain to the N-terminus of the human TGF RII or soluble fragment thereof capable of binding TGFp.
  • the polypeptide may include the amino acid sequence of SEQ ID NO: 3 or an amino acid sequence substantially identical to SEQ ID NO: 3.
  • the antibody fragment may be an scFv, Fab, F(ab') 2 , or Fv fragment.
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and human TGFpRII.
  • the antibody may optionally include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • a modified constant region e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain.
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment).
  • the antibody may optionally include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and a human TGF RII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and human
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and a fragment of human TGF RII capable of binding TGFP (e.g., a soluble fragment).
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal
  • Lys ⁇ Ala substitution a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and a human TGF RII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and human TGFpRII.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment).
  • the antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and a human TGF RII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu- Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and human TGFpRII.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment).
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and a human TGFpRII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and human TGFpRII.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and a fragment of human TGFpRII capable of binding TGF (e.g., a soluble fragment).
  • the antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and a human TGFpRII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu- Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and human TGFpRII.
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment).
  • the antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys- ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and a human TGFpRII ECD.
  • the antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys ⁇ Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
  • the invention also features a nucleic acid that includes a nucleotide sequence that encodes a polypeptide described above.
  • the nucleic acid further includes a second nucleotide sequence encoding at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-Ll (e.g., including amino acids 1-110 of SEQ ID NO: 1).
  • the second nucleotide sequence may encode the amino acid sequence of SEQ ID NO: 1 (secreted anti-PD-Ll lambda light chain) or an amino acid sequence substantially identical to SEQ ID NO: 1.
  • the invention also features a cell including any of the nucleic acids described above.
  • the invention also features a method of producing a protein including (a) the extracellular domain of the human TGFPRII, or a fragment thereof capable of binding TGFP (e.g., a soluble fragment), and (b) an antibody, or an antigen-binding fragment thereof, that binds human PD-Ll .
  • the method includes maintaining a cell described under conditions that permit expression of the protein.
  • the method may further include harvesting the protein.
  • the invention also features a protein including the polypeptide described above and at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-Ll.
  • the protein may include (a) two polypeptides, each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 3, and (b) two additional polypeptides each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 1.
  • the invention also features a protein described above for use in therapy.
  • the therapy may include administration of radiation or administration of a chemotherapeutic, a biologic, or a vaccine.
  • the invention also features a protein described above for use in promoting local depletion of TGFp at a tumor.
  • the invention also features a protein described above for use in inhibiting SMAD3 phosphorylation in a cell (e.g., a tumor cell or an immune cell). [0031] The invention also features a protein described above for use in treating cancer or for use in inhibiting tumor growth.
  • the cancer or tumor may be selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
  • the use may further include administration of radiation or administration of a chemotherapeutic, a biologic, or a vaccine.
  • the invention also features a method of promoting local depletion of TGFp.
  • the method includes administering a protein described above, where the protein binds TGFp in solution, binds PD-Ll on a cell surface, and carries the bound TGFp into the cell (e.g., a cancer cell).
  • the invention also features a method of inhibiting SMAD3 phosphorylation in a cell (e.g., a cancer cell or an immune cell), the method including exposing the cell in the tumor microenvironment to a protein described above.
  • a cell e.g., a cancer cell or an immune cell
  • the invention also features a method of inhibiting tumor growth or treating cancer.
  • the method includes exposing the tumor to a protein described above.
  • the method may further include exposing the tumor to radiation or to a chemotherapeutic, a biologic, or a vaccine.
  • the tumor or cancer is selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
  • TGFpRII or "TGF Receptor ⁇ ” is meant a polypeptide having the wild-type human TGFp Receptor Type 2 Isoform A sequence (e.g., the amino acid sequence of NCBI Reference Sequence (RefSeq) Accession No. NP_001020018 (SEQ ID NO: 8)), or a polypeptide having the wild-type human TGFP Receptor Type 2 Isoform B sequence (e.g., the amino acid sequence of NCBI RefSeq Accession No. NP 003233 (SEQ ID NO: 9)) or having a sequence substantially identical the amino acid sequence of SEQ ID NO: 8 or of SEQ ID NO: 9.
  • the TGF RII may retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGFP-binding activity of the wild-type sequence.
  • the polypeptide of expressed TGFpRII lacks the signal sequence.
  • fragment of TGFpRII capable of binding TGF is meant any portion of NCBI RefSeq Accession No. P_001020018 (SEQ ID NO: 8) or of NCBI RefSeq Accession No.
  • NP_003233 (SEQ ID NO: 9), or a sequence substantially identical to SEQ ID NO: 8 or SEQ ID NO: 9 that is at least 20 (e.g., at least 30, 40, 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 175, or 200) amino acids in length that retains at least some of the TGFp-binding activity (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%) of the wild-type receptor or of the corresponding wild-type fragment.
  • TGFp-binding activity e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%
  • TGFpRII extra-cellular domain having the sequence of SEQ ID NO: 10.
  • substantially identical is meant a polypeptide exhibiting at least 50%, desirably 60%, 70%, 75%, or 80%, more desirably 85%, 90%, or 95%, and most desirably 99% amino acid sequence identity to a reference amino acid sequence.
  • the length of comparison sequences will generally be at least 10 amino acids, desirably at least 15 contiguous amino acids, more desirably at least 20, 25, 50, 75, 90, 100, 150, 200, 250, 300, or 350 contiguous amino acids, and most desirably the full-length amino acid sequence.
  • patient is meant either a human or non-human animal (e.g., a mammal).
  • treating a disease, disorder, or condition (e.g., a cancer) in a patient is meant reducing at least one symptom of the disease, disorder, or condition by administrating a therapeutic agent to the patient.
  • cancer is meant a collection of cells multiplying in an abnormal manner.
  • FIG. 1A is a schematic drawing of an anti-PD-Ll /TGFp Trap molecule comprising one anti-PD-Ll antibody fused to two extracellular domain (ECD) of TGFP Receptor II via a (Gly 4 Ser) 4 Gly linker.
  • FIG. IB is a photograph of a sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of anti-PD-Ll/TGFp Trap under non-reducing and reducing conditions.
  • SDS-PAGE sodium dodecyl sulfate-polyacrylamide gel electrophoresis
  • FIG. 2 is photograph of an SDS-PAGE gel showing analysis of extent of clipping of anti-PD-Ll/TGF Trap expressed by clone 02B15 at various population doubling levels.
  • Anti- PD-Ll /TGFp Trap from clone 02B15 after a single protein A chromatography step was analyzed by SDS-PAGE under reducing conditions. Lanes 1 and 10, See Blue Plus 2 MW Standard; lane 2, purified anti-PD-Ll /TGFp Trap reference; lane 3, clone 02B15 at PDLO; lane 4, clone 02B15 at PDL30; lane 5, clone 02B 15 at PDL60; and lane 6, clone 02B15 at PDL90. (PDL, population doubling level).
  • FIG. 3 is a graph showing FACS analysis of anti-PD-Ll /TGFp Trap binding to HEK cells transfected to express human PD-L1.
  • FIG. 4 is a graph showing the ability of anti-PD-L 1/TGFP Trap to inhibit TGFp- induced phosphorylation of SMAD3 using a pSMAD3-luciferase reporter cell line (filled circle: anti-PD-Ll ; X: anti-PD-Ll (mut); filled square: anti-PD-Ll /TGFp Trap; filled triangle: anti-PD-Ll(mut)/TGFp Trap; +: anti-TGFp antibody 1D1 1 ; star: TGFp RII-Fc).
  • FIGS. 5A and 5B are graphs showing pharmacokinetics of intravenously administered anti-PD-Ll /TGFp Trap and related proteins in mice.
  • FIG. 6 A is a graph showing PD-L1 target-mediated endocytosis of anti-PD-Ll/ TGF Trap.
  • FIG. 6B is a graph showing PD-L1 target-mediated endocytosis of anti-PD-Ll .
  • FIG. 6C is a graph showing percent internalization of anti-PD-Ll/ TGFp Trap and anti-PD-Ll bound on HEK/PD-L 1 cells.
  • FIGS. 7A-7C are graphs showing anti-tumor efficacy of anti-PD-Ll /TGFp Trap and related proteins in the EMT-6 breast carcinoma subcutaneous model (Example 7).
  • FIG. 7A shows tumor growth curves of average tumor volumes of surviving mice in different treatment groups (star: Group 1 : filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; open square: Group 5; filled square/dashed line: Group 6; filled square/stippled line: Group 7).
  • FIG. 7B shows tumor growth curves of individual tumor volumes in different treatment groups.
  • FIG. 7C is a Kaplan-Meier plot of percent survival in different treatment groups (symbols as in 7A).
  • FIG. 8 is a graph showing anti-tumor efficacy of anti-PD-Ll /TGFp Trap and related proteins in the MC38 colorectal carcinoma subcutaneous tumor model (Example 8; star: Group 1; filled circle: Group 2; filled circle/dashed line: Group 3; filled triangle: Group 4; filled triangle/dashed line: Group 5; filled square: Group 6; filled square/dashed line: Group 7).
  • FIG. 9 is a graph showing anti-tumor efficacy of anti-PDLl/TGFp Trap and related proteins in an orthotopic EMT-6 breast cancer model (Example 9; star: Group 1; filled circle/dashed line: Group 2; filled triangle: Group 3; filled triangle/dashed line: Group 4; filled diamond: Group 5).
  • FIG. 10 is a graph showing anti-tumor efficacy of anti-PDLl/TGFp Trap and related proteins in an intramuscular MC38 colorectal carcinoma model (Example 10; star: Group 1; filled circle: Group 2; filled circle/dashed line: Group 3: filled diamond/dashed line: Group 4; filled square: Group 5; filled square/dashed line: Group 6; filled diamond: Group 7).
  • FIG. 11 is a graph showing anti-tumor efficacy of anti-PD-Ll/TGF- ⁇ Trap and the combination of anti-PD-Ll and TGF Trap control administered to give equivalent in vivo exposure in an orthotopic EMT-6 breast tumor model (Example 11 ; star: Group 1 ; filled square: Group 2; open square: Group 3; filled diamond: Group 4; open diamond: Group 5).
  • FIGS. 12A-12C are graphs showing anti-tumor efficacy of anti-PD-Ll /TGF-p Trap and the combination of anti-PD-Ll and TGF Trap control administered to give equivalent in vivo exposure in an intramuscular MC38 colorectal carcinoma model (Example 12).
  • FIG. 12 is a graph showing anti-tumor efficacy of anti-PD-Ll/TGF- ⁇ Trap and the combination of anti-PD-Ll and TGF Trap control administered to give equivalent in vivo exposure in an intramuscular MC38 colorectal carcinoma model
  • FIG. 12 A shows tumor growth curves of mice treated with both intermediate and low doses of the proteins (star: Group 1 ; filled squares: Group 2; open squares: Group 3; filled diamonds: Group 4; open diamonds Group 5).
  • FIG. 12B (star: Group I; filled square: Group 2; filled diamond: Group 4; *: p ⁇ 0.0001 compared to Group 1 ; **: p ⁇ 0.0001 compared to Group 2)
  • 12C (star: Group 1 ; filled square: Group3; filled diamond: Group 5; *: p ⁇ 0.0001 compared to Group 1 ; **: p ⁇ 0.0001 compared to Group 3) show statistical analysis of tumor growth curves of mice treated with intermediate and low doses of the proteins, respectively
  • FIGS. 13 A-13B are graphs showing anti-tumor efficacy of anti-PD-L 1 (YW)/TGF-p Trap and related proteins in an orthotopic EMT-6 breast tumor model (Example 13 ; star: Group 1; filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; filled diamond: Group 5).
  • FIG. 13A shows tumor growth curves of mice in different treatment groups.
  • FIG. 13B is a Kaplan-Meier plot of percent survival in different treatment groups.
  • FIGS. 14A-14B are graphs showing anti-tumor efficacy of anti-PD-Ll(YW)/TGF-p Trap and related proteins based on (A) tumor volumes and (B) tumor weights, in an intramuscular MC38 colorectal carcinoma model (Example 14; star: Group 1 ; filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; filled diamond: Group 5).
  • FIG. 15 is a graph comparing the anti-tumor efficacy of an anti-PD-1 antibody treatment with and without TGFp Trap control in an orthotopic EMT-6 breast tumor model (Example 15; star: Group 1 ; filled square: Group 2; filled inverted triangle: Group 3; open inverted triangle: Group 4).
  • FIG. 16 is a graph comparing the anti -tumor efficacy of an anti-PD-1 antibody treatment with and without TGFp Trap control in an intramuscular MC38 colorectal tumor model (Example 16; star: Group 1 ; filled square: Group 2; filled inverted triangle: Group 3; open inverted triangle: Group 4).
  • Example 18 is a graph comparing the anti-tumor efficacy of an anti-LAG3 or anti-TIM3 antibody treatment with and without TGFp Trap control in an intramuscular MC38 colorectal tumor model (Example 18; star: Group 1; filled square: Group 2; filled triangle: Group 3; filled inverted triangle: Group 4; open triangle: Group 5; open inverted triangle: Group 6).
  • the current invention permits localized reduction in TGF in a tumor
  • an antibody moiety of the invention to an immune checkpoint protein is anti-PD-Ll.
  • This bifunctional molecule sometimes referred to in this document as an "antibody-cytokine trap," is effective precisely because the anti -receptor antibody and cytokine trap are physically linked.
  • the resulting advantage (over, for example, administration of the antibody and the receptor as separate molecules) is partly because cytokines function predominantly in the local environment through autocrine and paracrine functions.
  • the antibody moiety directs the cytokine trap to the tumor microenvironment where it can be most effective, by neutralizing the local
  • Antibody-mediated target internalization has been shown for PD-L1. This is a distinct advantage over using an anti-TGFp antibody because first, an anti-TGFp antibody might not be completely neutralizing; and second, the antibody can act as a carrier extending the half-life of the cytokine, and antibody/cytokine complexes often act as a circulating sink that builds up and ultimately dissociates to release the cytokine back in circulation (Montero-Julian et al, Blood. 1995; 85:917-24).
  • cytokine trap to neutralize the ligand can also be a better strategy than blockading the receptor with an antibody, as in the case of CSF-1. Because CSF-1 is cleared from the circulation by receptor- mediated endocytosis, an anti-CSF-1 receptor antibody blockade caused a significant increase in circulating CSF-1 concentration (Hume et al, Blood. 2012;119: 1810-20)
  • anti-PD-Ll /TGFp Trap elicits a synergistic anti-tumor effect due to the simultaneous blockade of the interaction between PD- Ll on tumor cells and PD-1 on immune cells, and the neutralization of TGFP in the tumor microenvironment.
  • anti-PDLl/TGFp Trap has efficacy superior to that of the single agent anti-PD-Ll or TGFP Trap control. Without being bound by theory, this presumably is due to a synergistic effect obtained from simultaneous blocking the two major immune escape mechanisms, and in addition, the targeted depletion of the TGFp in the tumor microenvironment by a single molecular entity. This depletion is achieved by (1) anti-PD-Ll targeting of tumor cells; (2) binding of the TGFp
  • TGFP2 plays in embryonic development of the heart as well as in repair of myocardial damage after ischemia and reperfusion injury (Roberts et ah, J Clin Invest. 1992; 90:2056-62).
  • TGFP had been a somewhat questionable target in cancer immunotherapy because of its paradoxical roles as the molecular Jekyll and Hyde of cancer (Bierie et ah, Nat Rev Cancer. 2006; 6:506-20).
  • TGFp activity is developmental stage and context dependent. Indeed TGFp can act as either a tumor promoter or a tumor suppressor, affecting tumor initiation, progression and metastasis. The mechanisms underlying this dual role of TGFp remain unclear (Yang et ah, Trends Immunol. 2010; 31 :220-227).
  • TGFp ligand and the receptor have been studied intensively as therapeutic targets.
  • TGFpi ligand iso forms
  • TGFpR TGFP receptors
  • TGFpR type I, II and III TGFP receptors
  • TGFpRI is the signaling chain and cannot bind ligand.
  • TGFPRII binds the ligand TGFpi and 3, but not TGFp2, with high affinity.
  • TGFpRII/TGFp complex recruits TGFpRI to form the signaling complex (Won et ah, Cancer Res. 1999; 59:1273-7).
  • TGFpRIII is a positive regulator of TGFp binding to its signaling receptors and binds all 3 TGF isoforms with high affinity.
  • the TGFp/TGFpRJII complex binds TGFpRII and then recruits TGFpRI, which displaces
  • TGFpRIII to form the signaling complex.
  • TGFp isoforms all signal through the same receptor, they are known to have differential expression patterns and non-overlapping functions in vivo.
  • the three different TGF-p isoform knockout mice have distinct phenotypes, indicating numerous non-compensated functions (Bujak et ah, Cardiovasc Res. 2007; 74: 184-95). While TGF i null mice have hematopoiesis and vasculogenesis defects and TGFp3 null mice display pulmonary development and defective palatogenesis, TGFp2 null mice show various developmental abnormalities, the most prominent being multiple cardiac deformities (Bartram et al, Circulation. 2001; 103:2745-52; Yamagishi et al, Anat Rec. 2012; 295:257-67).
  • TGFP is implicated to play a major role in the repair of myocardial damage after ischemia and reperfusion injury.
  • cardiomyocytes secrete TGFp, which acts as an autocrine to maintain the spontaneous beating rate.
  • TGFp acts as an autocrine to maintain the spontaneous beating rate.
  • TGFP2 70-85% of the TGFP secreted by cardiomyocytes.
  • a therapeutic agent that neutralizes TGFpi but not TGFP2 could provide an optimal therapeutic index by minimizing the cardiotoxicity without compromising the anti-tumor activity. This is consistent with the findings by the present inventors, who observed a lack of toxicity, including cardiotoxicity, for anti-PD-Ll/TGFp Trap in monkeys.
  • Therapeutic approaches to neutralize TGFP include using the extracellular domains of TGFp receptors as soluble receptor traps and neutralizing antibodies.
  • soluble TGFpRIII may seem the obvious choice since it binds all the three TGFp ligands.
  • TGFpRIII which occurs naturally as a 280-330 kD glucosaminoglycan (GAG)-glycoprotein, with extracellular domain of 762 amino acid residues, is a very complex protein for biotherapeutic development.
  • the soluble TGFpRIII devoid of GAG could be produced in insect cells and shown to be a potent TGFp neutralizing agent (Vilchis-Landeros et al, Biochem J 355:215, 2001).
  • the two separate binding domains (the endoglin-related and the uromodulin-related) of TGFpRIII could be independently expressed, but they were shown to have affinities 20 to 100 times lower than that of the soluble TGFPRIII, and much diminished neutralizing activity (Mendoza et al, Biochemistry. 2009; 48: 11755-65).
  • the extracellular domain of TGFpRII is only 136 amino acid residues in length and can be produced as a glycosylated protein of 25-35 kD.
  • the recombinant soluble TGFPRII was further shown to bind TGFpi with a D of 200 pM, which is fairly similar to the KD of 50 pM for the full length TGFpRII on cells (Lin et al, J Biol Chem. 1995; 270:2747-54). Soluble TGFpRII- Fc was tested as an anti-cancer agent and was shown to inhibit established murine malignant mesothelioma growth in a tumor model (Suzuki et al, Clin Cancer Res. 2004; 10:5907-18).
  • TGFpRII does not bind TGFp2
  • TGFpRIII binds TGFpi and 3 with lower affinity than TGFpRII
  • a fusion protein of the endoglin domain of TGFpRIII and extracellular domain of TGFpRII was produced in bacteria and was shown to inhibit the signaling of TGFpi and 2 in cell based assays more effectively than either TGFpRII or RIII (Verona et al, Protein Eng Des Sel. 2008; 21 :463-73).
  • TGFp receptor trap recombinant proteins have been tested in the clinic.
  • Still another approach to neutralize all three isoforms of the TGFp ligands is to screen for a pan-neutralizing anti-TGFp antibody, or an anti-receptor antibody that blocks the receptor from binding to TGFpi, 2 and 3.
  • GC1008 a human antibody specific for all isoforms of TGFp, was in a Phase I/II study in patients with advanced malignant melanoma or renal cell carcinoma (Morris et al, J Clin Oncol 2008; 26:9028 (Meeting abstract)).
  • Metelimumab an antibody specific for TGFpi was tested in Phase 2 clinical trial as a treatment to prevent excessive post-operative scarring for glaucoma surgery; and Lerdelimumab, an antibody specific for TGFP2, was found to be safe but ineffective at improving scarring after eye surgery in a Phase 3 study (Khaw et al , Ophthalmology 2007; 114: 1822-1830).
  • Anti-TGFpRII antibodies that block the receptor from binding to all three TGFP isoforms such as the anti- human TGFPRII antibody TRl and anti-mouse TGFpRII antibody MTl, have also shown some therapeutic efficacy against primary tumor growth and metastasis in mouse models (Zhong et al, Clin Cancer Res.
  • the antibody-TGFp trap of the invention is a bifunctional protein containing at least portion of a human TGFP Receptor II (TGFpRII) that is capable of binding TGFp.
  • TGFpRII human TGFP Receptor II
  • the TGF trap polypeptide is a soluble portion of the human TGF Receptor Type 2 Isoform A (SEQ ID NO: 8) that is capable of binding TGFp.
  • TGFp trap polypeptide contains at least amino acids 73-184 of SEQ ID NO:8.
  • the TGFp trap polypeptide contains amino acids 24-184 of SEQ ID NO:8.
  • the TGFp trap polypeptide is a soluble portion of the human TGFp Receptor Type 2 Isoform B (SEQ ID NO: 9) that is capable of binding TGFp.
  • TGFp trap polypeptide contains at least amino acids 48-159 of SEQ ID NO:9.
  • the TGFP trap polypeptide contains amino acids 24-159 of SEQ ID NO:9.
  • the TGFp trap polypeptide contains amino acids 24-105 of SEQ ID NO:9.
  • T cell inhibition checkpoints for dis-inhibition with therapeutic antibodies is an area of intense investigation (for a review, see Pardoll, Nat Rev Cancer. 2012; 12:253-264).
  • the antibody moiety or antigen binding fragment thereof targets T cell inhibition checkpoint receptor proteins on the T cell, such as, for example: CTLA-4, PD-1, BTLA, LAG-3, TIM-3, and LAIR1.
  • the antibody moiety targets the counter-receptors on antigen presenting cells and tumor cells (which co-opt some of these counter-receptors for their own immune evasion), such as, for example: PD-Ll (B7-H1), B7-DC, HVEM, TIM-4, B7-H3, or B7-H4.
  • the invention contemplates antibody TGFp traps that target, through their antibody moiety or antigen binding fragment thereof, T cell inhibition checkpoints for dis-inhibition.
  • TGFp traps that target, through their antibody moiety or antigen binding fragment thereof, T cell inhibition checkpoints for dis-inhibition.
  • the present inventors have tested the anti-tumor efficacy of combining a TGFp trap with antibodies targeting various T cell inhibition checkpoint receptor proteins, such as anti- PD-1, anti-PD-Ll, anti -TIM-3 and anti-LAG3.
  • the experimental results are further detailed in Examples 7-18.
  • the present inventors found that combining a TGFP trap with an anti-PD-Ll antibody exhibited remarkable anti-tumor activity beyond what was observed with the monotherapies. In contrast, none of the other combinations with antibodies to the targets listed above showed any superior efficacy.
  • a combination treatment of a TGFp trap with an anti-PD-1 antibody would demonstrate similar activity to the one observed with anti-PD-Ll, as PD-1 / PD-Ll are cognate receptors that bind to each other to effect the immune checkpoint inhibition.
  • this is not what the present inventors have found.
  • the invention can include any anti-PD-Ll antibody, or antigen-binding fragment thereof, described in the art.
  • Anti-PD-Ll antibodies are commercially available, for example, the 29E2A3 antibody (Biolegend, Cat. No. 329701).
  • Antibodies can be monoclonal, chimeric, humanized, or human.
  • Antibody fragments include Fab, F(ab')2, scFv and Fv fragments, which are described in further detail below.
  • Exemplary antibodies are described in PCT Publication WO 2013/079174. These antibodies can include a heavy chain variable region polypeptide including an HVR-Hl , HVR- Hl, and HVR-H3 sequence, where:
  • the HVR-Hl sequence is X1YX2MX3;
  • the HVR-H2 sequence is SIYPSGGX 4 TFYADX 5 VKG;
  • the HVR-H3 sequence is IKLGTVTTVX 6 Y; further where: Xi is , R, T, Q, G, A, W, M, I, or S; X2 is V, R, K, L, M, or I; X 3 is H, T, N, Q, A, V, Y, W, F, or M; X 4 is F or I; X 5 is S or T; X 6 is E or D.
  • Xi is M, I, or S;
  • X 2 is L, M, or I;
  • X3 is F or M;
  • X 4 is I;
  • X5 is S or T;
  • X6 is D.
  • Xi is S; X 2 is I; X 3 is M; X 4 is I; X5 is T; X 6 is D.
  • the polypeptide further includes variable region heavy chain framework sequences juxtaposed between the HVRs according to the formula: (HC-FRl )- (HVR-Hl )-(HC-FR2)-(HVR-H2)-(HC-FR3)-(HVR-H3)-(HC-FR4).
  • the framework sequences are derived from human consensus framework sequences or human germline framework sequences.
  • HC-FRl is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
  • HC-FR2 is W VRQ APGKGLE WVS ;
  • HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
  • HC-FR4 is WGQGTLVTVSS.
  • the heavy chain polypeptide is further combined with a variable region light chain including an HVR-L1 , HVR-L2, and HVR-L3, where: (a) the HVR-L1 sequence is TGTX7X8DVGX9YNYVS;
  • the HVR-L3 sequence is SSX13TX14X15X16X17RV; further where: X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; Xn is I, N or S; X12 is D, H or N; X13 is F or Y; Xn is N or S; X15 is R, T or S; Xi 6 is G or S; X17 is I or T.
  • X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; X11 is N or S; X12 is N; X13 is F or Y; XH is S; Xis is S; Xi6 is G or S; X17 is T.
  • the light chain further includes variable region light chain framework sequences juxtaposed between the HVRs according to the formula: (LC- FR1MHVR-L1)-(LC-FR2)-(HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
  • the light chain framework sequences are derived from human consensus framework sequences or human germline framework sequences. [0083] In a still further aspect, the light chain framework sequences are lambda light chain sequences.
  • At least one of the framework sequence is the following:
  • LC-FR1 is QSALTQPASVSGSPGQSITISC;
  • LC-FR2 is WYQQHPGKAP LMIY
  • LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC
  • LC-FR4 is FGTGTKVTVL.
  • the invention provides an anti-PD-Ll antibody or antigen binding fragment including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain includes an HVR-Hl, HVR-H2, and HVR-H3, wherein further: (i) the HVR-Hl sequence is X1YX2MX 3 ; (ii) the HVR-H2 sequence is
  • the HVR-H3 sequence is IKLGTVTTVX 6 Y, and;
  • the light chain includes an HVR-L1, HVR-L2, and HVR-L3, wherein further: (iv) the HVR-L1 sequence is TGTX7X8DVGX9YNYVS; (v) the HVR-L2 sequence is
  • the HVR-L3 sequence is SSXi3TXi 4 Xi 5 Xi 6 XnRV; wherein: Xi is K, R, T, Q, G, A, W, M, I, or S; X2 is V, R, K, L, M, or I; X 3 is H, T, N, Q, A, V, Y, W, F, or M; X 4 is F or I; X5 is S or T; X 6 is E or D; X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; X11 is I, N, or S; X12 is D, H, or N; Xi 3 is F or Y; X14 is N or S; X15 is R, T, or S; Xi 6 is G or S; Xi7 is I or T.
  • Xi is M, I, or S;
  • X 2 is R, K, L, M, or I;
  • X is F or M;
  • X 4 is F or I;
  • X is S or T;
  • X 6 is E or D;
  • X7 is N or S;
  • Xs is T, R, or S;
  • X9 is A or G;
  • Xio is E or D;
  • X12 is N;
  • X13 is F or Y;
  • Xu is S;
  • X15 is S;
  • Xie is G or S;
  • X17 is T.
  • Xi is M, I, or S; X 2 is L, M, or I; X3 is F or M; X 4 is I; Xs is S or T; X 6 is D; X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; Xi 1 is N or S; X12 is N; Xn is F or Y; Xi 4 is S; Xis is S; Xi 6 is G or S; Xn is T.
  • the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1 MHVR-Ll )-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
  • the framework sequences are derived from human consensus framework sequences or human germline sequences.
  • one or more of the heavy chain framework sequences is the following: HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
  • HC-FR2 is WVRQAPGKGLEWVS
  • HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
  • HC-FR4 is WGQGTLVTVSS.
  • the light chain framework sequences are lambda light chain sequences.
  • one or more of the light chain framework sequences is the following: LC-FR1 is QSALTQPASVSGSPGQSITISC;
  • LC-FR2 is WYQQHPGKAPKLMIY
  • LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC;
  • LC-FR4 is FGTGTKVTVL.
  • the heavy chain variable region polypeptide, antibody, or antibody fragment further includes at least a CHI domain.
  • the heavy chain variable region polypeptide, antibody, or antibody fragment further includes a CHI, a CH2, and a CH3 domain.
  • variable region light chain, antibody, or antibody fragment further includes a CL domain.
  • the antibody further includes a CHI, a CH2, a CH3, and a CL domain.
  • the antibody further includes a human or murine constant region.
  • the human constant region is selected from the group consisting of lgGl , IgG2, IgG2, IgG3, IgG4.
  • the human or murine constant region is lgGl.
  • the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain includes an HVR-Hl, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to SYIMM, SIYPSGGITFYADTVKG, and IKLGTVTTVDY, respectively
  • the light chain includes an HVR-Ll, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTS SD VGGYNY VS , DVSNRPS, and SSYTSSSTRV, respectively.
  • sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain includes an HVR-Hl, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to MYMMM, SIYPSGGITFYADS VKG, and IKLGTVTTVDY, respectively, and
  • the light chain includes an HVR-Ll, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTSSDVGAYNYVS, DVSNRPS, and SSYTSSSTRV, respectively.
  • sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1)-(HVR-L1)-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
  • the framework sequences are derived from human germline sequences.
  • HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
  • HC-FR2 is WVRQ APGKGLE WVS ;
  • HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
  • HC-FR4 is WGQGTLVTVSS.
  • the light chain framework sequences are derived from a lambda light chain sequence.
  • one or more of the light chain framework sequences is the following:
  • LC-FR1 is QSALTQPASVSGSPGQSITISC;
  • LC-FR2 is WYQQHPGKAPKLMIY
  • LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC
  • LC-FR4 is FGTGTKVTVL.
  • the antibody further includes a human or murine constant region.
  • the human constant region is selected from the group consisting of IgG 1 , IgG2, IgG2, IgG3 , IgG4.
  • the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYIMMVWRQAPGKGLEWVSSIYPSGGITF YADWKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR1KLGTVTTVDYWGQGTLV TVSS, and
  • the light chain sequence has at least 85% sequence identity to the light chain sequence: QS ALTQPAS VSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSN RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGTKVTVL.
  • sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the invention provides for an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain sequence has at least 85 % sequence identity to the heavy chain sequence:
  • the light chain sequence has at least 85% sequence identity to the light chain sequence:
  • sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%.
  • the antibody binds to human, mouse, or cynomolgus monkey PD-Ll .
  • the antibody is capable of blocking the interaction between human, mouse, or cynomolgus monkey PD-Ll and the respective human, mouse, or cynomolgus monkey PD-1 receptors.
  • the antibody binds to human PD-Ll with a KD of 5xl0 "9 M or less, preferably with a KD of 2xl0 "9 M or less, and even more preferred with a KD of lxlO "9 M or less.
  • the invention relates to an anti-PD-Ll antibody or antigen binding fragment thereof which binds to a functional epitope including residues Y56 and D61 of human PD-Ll.
  • the functional epitope further includes E58, E60, Q66, Rl 13, and Ml 15 of human PD-Ll.
  • the antibody binds to a conformational epitope, including residues 54-66 and 112-122 of human PD-Ll.
  • the invention is related to an anti-PD-Ll antibody, or antigen binding fragment thereof, which cross-competes for binding to PD-Ll with an antibody according to the invention as described herein.
  • the invention features proteins and polypeptides including any of the above described anti-PD-Ll antibodies in combination with at least one pharmaceutically acceptable carrier.
  • the invention features an isolated nucleic acid encoding a polypeptide, or light chain or a heavy chain variable region sequence of an anti-PD-Ll antibody, or antigen binding fragment thereof, as described herein.
  • the invention provides for an isolated nucleic acid encoding a light chain or a heavy chain variable region sequence of an anti-PD-Ll antibody, wherein:
  • the heavy chain includes an HVR-H1, an HVR-H2, and an HVR-H3 sequence having at least 80% sequence identity to SYIMM, SIYPSGGITFYADTVKG, and
  • the light chain includes an HVR-L1, an HVR-L2, and an HVR-L3 sequence having at least 80% sequence identity to TGTSSDVGGYNYVS, DVSNRPS, and SSYTSSSTRV, respectively.
  • sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • nucleic acid sequence for the heavy chain is: atggagttgc ctgttaggct gttggtgctg atgttctgga ttcctgctag ctccagcgag 60
  • gacaccgtga agggccggtt caccatctcc cgggacaact ccaagaacac cctgtacctg 300
  • aagagcctct ccctgtcccc gggtaa 1407 and the nucleic acid sequence for the light chain is: atggagttgc ctgttaggct gttggtgctg atgttctgga ttcctgcttc cttaagccag 60 tccgcctga cccagcctgc ctgtgtct ggctccctg gccagtccat caccatcagc 120
  • anti-PD-Ll antibodies that can be used in an anti-PD-Ll TGFp Trap are described in US patent application publication US 2010/0203056.
  • the antibody moiety is YW243.55S70.
  • the antibody moiety is MPDL3280A.
  • the invention features an anti-PD-Ll antibody moiety including a heavy chain and a light chain variable region sequence, where:
  • the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWIHWVRQAPGKGLEWVAWISPYGGSTYY
  • the light chain sequence has at least 85% sequence identity to the light chain sequence: DIQMTQSPSSLSASVGDRVTITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLTISSLQPEDFATYYCQQYLYHPATFGQGTKVEIKR (SEQ ID NO: 13).
  • sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • the invention features an anti-PD-Ll antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain variable region sequence is:
  • the invention features an anti-PD-L 1 antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain variable region sequence is:
  • the light chain variable region sequence is: DIQMTQS PSSLSASVGDRVTITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLTI S SLQPEDFATYYCQQYLYHPATFGQGTKVE IKR (SEQ ID NO:13).
  • the anti-PD-L 1 antibody is MDX-1 105.
  • the anti-PD-Ll antibody is MEDI-4736.
  • the proteins and peptides of the invention can include a constant region of an immunoglobulin or a fragment, analog, variant, mutant, or derivative of the constant region.
  • the constant region is derived from a human immunoglobulin heavy chain, for example, IgGl , IgG2, IgG3, IgG4, or other classes.
  • the constant region includes a CH2 domain.
  • the constant region includes CH2 and CH3 domains or includes hinge-CH2-CH3.
  • the constant region can include all or a portion of the hinge region, the CH2 domain and/or the CH3 domain.
  • the constant region contains a mutation that reduces affinity for an Fc receptor or reduces Fc effector function.
  • the constant region can contain a mutation that eliminates the glycosylation site within the constant region of an IgG heavy chain.
  • the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu234, Leu235, Gly236, Gly237, Asn297, or Pro331 of IgGl (amino acids are numbered according to EU nomenclature).
  • the constant region contains a mutation at an amino acid position corresponding to Asn297 of IgGl .
  • the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu281, Leu282, Gly283, Gly284, Asn344, or Pro378 of IgGl .
  • the constant region contains a CH2 domain derived from a human IgG2 or IgG4 heavy chain.
  • the CH2 domain contains a mutation that eliminates the glycosylation site within the CH2 domain.
  • the mutation alters the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the CH2 domain of the IgG2 or lgG4 heavy chain.
  • the mutation changes the asparagine to a glutamine.
  • the mutation alters both the phenylalanine and the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence.
  • the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence is replaced with a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence.
  • the asparagine within the Gln-Phe- Asn-Ser (SEQ ID NO: 15) amino acid sequence corresponds to Asn297 of IgGl.
  • the constant region includes a CH2 domain and at least a portion of a hinge region.
  • the hinge region can be derived from an immunoglobulin heavy chain, e.g., IgGl, IgG2, IgG3, IgG4, or other classes.
  • the hinge region is derived from human IgGl, IgG2, IgG3, IgG4, or other suitable classes. More preferably the hinge region is derived from a human IgGl heavy chain.
  • the cysteine in the Pro- Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence of the IgGl hinge region is altered.
  • the Pro-Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence is replaced with a Pro-Lys-Ser-Ser-Asp-Lys (SEQ ID NO: 18) amino acid sequence.
  • the constant region includes a CH2 domain derived from a first antibody isotype and a hinge region derived from a second antibody isotype.
  • the CH2 domain is derived from a human IgG2 or IgG4 heavy chain, while the hinge region is derived from an altered human IgGl heavy chain.
  • junction region of a protein or polypeptide of the present invention can contain alterations that, relative to the naturally-occurring sequences of an immunoglobulin heavy chain and
  • the constant region is derived from an IgG sequence in which the C-terminal lysine residue is replaced.
  • the C-terminal lysine of an IgG sequence is replaced with a non-lysine amino acid, such as alanine or leucine, to further increase serum half-life.
  • the constant region is derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence near the C-terminus of the constant region is altered to eliminate potential junctional T-cell epitopes.
  • the Leu-Ser-Leu-Ser amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence.
  • the amino acids within the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment are replaced with other amino acids such as glycine or proline.
  • Detailed methods of generating amino acid substitutions of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment near the C- terminus of an IgGl, IgG2, IgG3, IgG4, or other immunoglobulin class molecule have been described in U.S. Patent Publication No. 2003/0166877, the disclosure of which is hereby incorporated by reference.
  • Suitable hinge regions for the present invention can be derived from IgGl, IgG2, IgG3, IgG4, and other immunoglobulin classes.
  • the IgGl hinge region has three cysteines, two of which are involved in disulfide bonds between the two heavy chains of the immunoglobulin. These same cysteines permit efficient and consistent disulfide bonding formation between Fc portions. Therefore, a preferred hinge region of the present invention is derived from IgGl, more preferably from human IgGl.
  • the first cysteine within the human IgGl hinge region is mutated to another amino acid, preferably serine.
  • the IgG2 isotype hinge region has four disulfide bonds that tend to promote oligomerization and possibly incorrect disulfide bonding during secretion in recombinant systems.
  • a suitable hinge region can be derived from an IgG2 hinge; the first two cysteines are each preferably mutated to another amino acid.
  • the hinge region of IgG4 is known to form interchain disulfide bonds inefficiently.
  • a suitable hinge region for the present invention can be derived from the IgG4 hinge region, preferably containing a mutation that enhances correct formation of disulfide bonds between heavy chain-derived moieties (Angal S, et al. (1993) Mol. Immunol., 30: 105-8).
  • the constant region can contain CH2 and/or CH3 domains and a hinge region that are derived from different antibody isotypes, i.e., a hybrid constant region.
  • the constant region contains CH2 and/or CH3 domains derived from IgG2 or IgG4 and a mutant hinge region derived from IgGl.
  • a mutant hinge region from another IgG subclass is used in a hybrid constant region.
  • a mutant form of the IgG4 hinge that allows efficient disulfide bonding between the two heavy chains can be used.
  • a mutant hinge can also be derived from an IgG2 hinge in which the first two cysteines are each mutated to another amino acid. Assembly of such hybrid constant regions has been described in U.S. Patent Publication No. 2003/0044423, the disclosure of which is hereby incorporated by reference.
  • the constant region can contain one or more mutations described herein.
  • the combinations of mutations in the Fc portion can have additive or synergistic effects on the prolonged serum half-life and increased in vivo potency of the bifunctional molecule.
  • the constant region can contain (i) a region derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence; (ii) a C-terminal alanine residue instead of lysine; (iii) a CH2 domain and a hinge region that are derived from different antibody isotypes, for example, an IgG2 CH2 domain and an altered IgGl hinge region; and (iv) a mutation that eliminates the glycosylation site within the IgG2-derived CH2 domain, for example, a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence instead of the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the IgG2-derived CH2 domain.
  • the proteins and polypeptides of the invention can also include antigen-binding fragments of antibodies.
  • Exemplary antibody fragments include scFv, Fv, Fab, F(ab') 2 , and single domain VHH fragments such as those of camelid origin.
  • Single-chain antibody fragments also known as single-chain antibodies (scFvs) are recombinant polypeptides which typically bind antigens or receptors; these fragments contain at least one fragment of an antibody variable heavy-chain amino acid sequence (VH) tethered to at least one fragment of an antibody variable light-chain sequence (VL) with or without one or more interconnecting linkers.
  • VH antibody variable heavy-chain amino acid sequence
  • VL variable light-chain sequence
  • Such a linker may be a short, flexible peptide selected to assure that the proper three-dimensional folding of the VL and VH domains occurs once they are linked so as to maintain the target molecule binding-specificity of the whole antibody from which the single-chain antibody fragment is derived.
  • the carboxyl terminus of the VL or VH sequence is covalently linked by such a peptide linker to the amino acid terminus of a complementary VL and VH sequence.
  • Single-chain antibody fragments can be generated by molecular cloning, antibody phage display library or similar techniques. These proteins can be produced either in eukaryotic cells or prokaryotic cells, including bacteria.
  • Single-chain antibody fragments contain amino acid sequences having at least one of the variable regions or CDRs of the whole antibodies described in this specification, but are lacking some or all of the constant domains of those antibodies. These constant domains are not necessary for antigen binding, but constitute a major portion of the structure of whole antibodies. Single-chain antibody fragments may therefore overcome some of the problems associated with the use of antibodies containing part or all of a constant domain. For example, single-chain antibody fragments tend to be free of undesired interactions between biological molecules and the heavy-chain constant region, or other unwanted biological activity.
  • single-chain antibody fragments are considerably smaller than whole antibodies and may therefore have greater capillary permeability than whole antibodies, allowing single- chain antibody fragments to localize and bind to target antigen-binding sites more efficiently.
  • antibody fragments can be produced on a relatively large scale in prokaryotic cells, thus facilitating their production.
  • the relatively small size of single-chain antibody fragments makes them less likely than whole antibodies to provoke an immune response in a recipient.
  • Fragments of antibodies that have the same or comparable binding characteristics to those of the whole antibody may also be present. Such fragments may contain one or both Fab fragments or the F(ab') 2 fragment.
  • the antibody fragments may contain all six CDRs of the whole antibody, although fragments containing fewer than all of such regions, such as three, four or five CDRs, are also functional. Protein production
  • the antibody-cytokine trap proteins are generally produced recombinantly, using mammalian cells containing a nucleic acid engineered to express the protein. Although one example of a suitable cell line and protein production method is described in Examples 1 and 2, a wide variety of suitable vectors, cell lines and protein production methods have been used to produce antibody-based biopharmaceuticals and could be used in the synthesis of these antibody-cytokine trap proteins.. Therapeutic indications
  • the anti-PD-Ll/TGF Trap proteins described in the application can be used to treat cancer or reduce tumor growth in a patient.
  • Exemplary cancers include colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
  • the cancer or tumor to be treated with an anti-PD-Ll/ TGFp Trap may be selected based on the expression or elevated expression of PD-L1 and TGFP in the tumor, the correlation of their expression levels with prognosis or disease progression, and preclinical and clinical experience on the sensitivity of the tumor to treatments targeting PD-L1 and TGFp.
  • Such cancers or tumors include but are not limited to colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, bladder, head and neck, liver, non-small cell lung cancer, melanoma, Merkel cell carcinoma, and mesothelioma.
  • the present invention also features pharmaceutical compositions that contain a therapeutically effective amount of a protein described herein.
  • the composition can be formulated for use in a variety of drug delivery systems.
  • One or more physiologically acceptable excipients or carriers can also be included in the composition for proper
  • the pharmaceutical compositions are intended for parenteral, intranasal, topical, oral, or local administration, such as by a transdermal means, for therapeutic treatment.
  • the pharmaceutical compositions can be administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion, or by topical application or intraarticular injection at areas affected by the vascular or cancer condition. Additional routes of administration include intravascular, intra-arterial, intratumor, intraperitoneal,
  • compositions for parenteral administration that comprise the above mention agents dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, PBS, and the like.
  • an acceptable carrier preferably an aqueous carrier
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents and the like.
  • compositions for oral delivery which may contain inert ingredients such as binders or fillers for the formulation of a tablet, a capsule, and the like.
  • compositions for local administration which may contain inert ingredients such as solvents or emulsifiers for the formulation of a cream, an ointment, and the like.
  • compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as-is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 1 1 , more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5.
  • the resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules.
  • the composition in solid form can also be packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.
  • the optimal dose of the antibody-TGFP trap is based on the percent receptor occupancy by the antibody moiety to achieve maximal therapeutic effect because the cytokine trap is used in a large excess.
  • the therapeutic dose for a monoclonal antibody targeting a cellular receptor is determined such that the trough level is around 10 to 100 ⁇ g/ml, i.e., 60 to 600 nM (for antibody with a dissociation constant (KD) of 6 nM, this trough level would ensure that between 90 to 99% of the target receptors on the cells are occupied by the antibody).
  • KD dissociation constant
  • the optimal dose of antibody-TGFP trap polypeptide of the invention will depend on the disease being treated, the severity of the disease, and the existence of side effects.
  • the optimal dose can be determined by routine experimentation.
  • a dose between 0.1 mg/kg and 100 mg/kg, alternatively between 0.5 mg/kg and 50 mg/kg, alternatively, between 1 mg/kg and 25 mg/kg, alternatively between 2 mg/kg and 10 mg/kg, alternatively between 5 mg/kg and 10 mg/kg is administered and may be given, for example, once weekly, once every other week, once every third week, or once monthly per treatment cycle.
  • Anti-PD-Ll /TGFp Trap is an anti-PD-Ll antibody-TGFp Receptor II fusion protein.
  • the light chain of the molecule is identical to the light chain of the anti-PD-Ll antibody (SEQ ID NO: 1).
  • the heavy chain of the molecule (SEQ ID NO:3) is a fusion protein comprising the heavy chain of the anti-PD-Ll antibody (SEQ ID NO: 2) genetically fused to via a flexible
  • the purified protein comprising one anti-PD-Ll antibody and two soluble TGFB Receptor II molecules (FIG. 1A) has an estimated molecular weight (MW) of about 190 kilodaltons on size exclusion chromatography and SDS-polyacrylamide electrophoresis under non-reducing conditions. Under reducing conditions, the light and heavy chains have apparent MW of 28 and 75 kilodaltons, respectively (FIG. IB).
  • the anti-PD-Ll (mut)/TGFp Trap fusion protein which contains an analogous heavy chain fusion polypeptide (SEQ ID NO:7) and a light chain with the mutations A31G, D52E, R99Y in the variable region that abrogate the binding to PD-L1 (SEQ ID NO:6), was similarly prepared. It was used in subsequent experiments as a TGFP Trap control.
  • the anti-PD-Ll/TGFp Trap produced by transient transfection of human embryonic kidney 293 (HEK) cells was found to contain varying degrees of a clipped species, which appeared as a faint band with an apparent MW of about 60 kD on SDS-PAGE under reducing conditions (FIG. IB). This band was confirmed to be the heavy chain of the anti-PD-Ll/TGFp Trap cleaved at a site in the N-terminal portion of TGFpRII close to the fusion junction.
  • Stable clones expressing anti-PD-Ll/TGFP Trap were generated in the CHO-S host cell line, which was pre-adapted for growth in serum-free media in suspension culture.
  • Cells were transfected with an expression vector containing a gene encoding the anti-PD-Ll- TGFpRII protein and a glutamine synthetase selection marker. Subsequent selection of stable integrants was made with L-methionine sulfoximine (MSX).
  • MSX L-methionine sulfoximine
  • Anti-PD-Ll/TGF Trap expressing cell lines were generated using a minipool approach, followed by the deposition of single cells into 384-well plates, using a Beckton-Dickinson fluorescence activated cell sorter (FACS Aria II).
  • HEK cells stably transfected to express human PD-Ll at 5 xlO 6 cells/ml were dispensed to the wells with test samples and mixed.
  • EC50 was calculated using non-linear regression (Sigmoidal dose-response) with Graphpad Prism5.
  • Fig. 3 FACS analysis showed that the anti-PD-Ll /TGFp Trap fusion protein retains similar binding affinity as the positive control anti-PD-Ll antibody on HEK cells stably transfected to express human PD-Ll (HEK/PD-Ll cells).
  • the EC50's for anti-PD- Ll/TGF Trap and anti-PD-Ll are 0.1 16 ⁇ (0.64 nM) and 0.061 ⁇ (0.41 nM), respectively.
  • the observed MFI (mean fluorescent intensity) was specific to binding to human PD-Ll since no MFI was observed on the parental HEK cells that were not transfected.
  • the anti-PD-Ll (mut)/TGF Trap negative control did not show any binding to the HEK cells stably transfected to express human PD-Ll.
  • Luminescence was measured using Envision 2104 plate reader by recording CPM.
  • Inhibition (%) (1- CPM of sample / CPM max of anti-PD-Ll treated sample) X 100
  • Nonlinear regression fit was carried out using Sigmoidal dose-response (variable slope) of Graphpad prism 5. IC50 values were calculated.
  • FIG. 4 shows that anti-PD-L l/TGFp Trap inhibits TGFp-induced pSMAD3 reporter activity in a dose dependent manner.
  • anti-PD-L l /TGFp Trap was several-fold more potent than TGFpRII-Fc (R&D Systems), which places the TGFPRII at the N-terminus instead of the C-terminus of the fusion protein.
  • anti-PD-Ll /TGFp Trap is significantly more potent than IDl 1 (GC1008), the anti-TGFp antibody that was tested in patients with advanced malignant melanoma or renal cell carcinoma (Morris et ah, J Clin Oncol 2008; 26:9028 (Meeting abstract)).
  • IDl 1 and TGFpRII-Fc showed similar activity.
  • EXAMPLE 5 Pharmacokinetic (PK) analysis in mice
  • Mouse body weight was recorded before dosing. After a brief warm-up under a heating lamp, each mouse received 120 ⁇ g of protein in 200 ⁇ intravenously (IV) via the tail vein regardless of its body weight.
  • IV intravenously
  • one subgroup was withdrawn for blood samples at lh, 24h, 72h, and 168h, whereas another subgroup was for blood samples at 7h, 48h, 120h, and 240h.
  • approximate 50 ⁇ of blood samples were collected from each mouse via tail vein using a heparinized micro glass capillary (100 ⁇ in capacity).
  • the blood sample was then transferred to a tube pre-coated with Li-Heparin and kept at 4 °C.
  • the blood samples were spun at 14,000 rpm for 10 min. At least 20 ⁇ of plasma sample was transferred into a new set of pre-labeled tubes and stored at -20 °C until the day of analysis.
  • the ELISA to measure fully functional anti-PD-Ll antibody and/or fusion protein used PD-Ll-Fc (extracellular domain of human PD-L1 fused to Fc) coated wells (coated at 1.25 ⁇ g/ml) for capture and peroxidase-AffiniPure mouse anti-Human IgG, F(ab')2 for detection.
  • the ELISA to measure fully functional anti-PD-Ll and intact TGFpRII used PD- Ll-Fc coated wells for capture and biotinylated anti -human TGF RII (R&D Systems) for detection.
  • FIG. 5A shows that the anti-PD-Ll /TGFp Trap fusion protein had a PK profile very similar to that of the anti-PD-Ll antibody.
  • the serum concentrations at the 168 hr time point of anti-PD-Ll /TGFp Trap and anti- PD-Ll were 16.8 and 16.2 ⁇ g/ml, respectively, and the respective area under the curve (AUC) from 0 to 168 hr were 4102 and 3841 hr ⁇ g/ml.
  • the serum concentrations at the 168 hr time point of anti-PD-Ll /TGFp Trap and anti-PD-Ll were 9.5 and 1 1.1 ⁇ g/ml, respectively, and the respective AUC from 0 to 168 hr were 3562 and 3086 hr ⁇ g/ml.
  • concentration of intact anti-PD-Ll/TGFp Trap fusion protein was determined by the ELISA, which detects fully functional anti-PD-Ll and the fused TGF RII.
  • the serum concentration of anti-PD-Ll/TGFP Trap was 5.9 ⁇ g/ml at the 168 hr time point and the AUC (0 to 168 hr) was 2656 hr- ⁇ g/ml, which were somewhat lower than those from the fully functional anti-PD-Ll ELISA, presumably due to degradation of the TGFpRII moiety after receptor- mediated endocytosis.
  • Antibody binding to PD-Ll has been shown to result in PD-Ll -mediated endocytosis, and an antibody-X fusion protein is known to undergo degradation of the X moiety after receptor-mediated endocytosis (Gillies et ah, Clin Cancer Res. 2002; 8:210-6). This is supported by the finding in FIG.
  • the exposure is about 3 times higher, with a serum concentration of 53 ⁇ g/ml at the 168 hr time point and AUC(0 to 168 hr) of 9585 hr- ⁇ g/ml, suggesting that at least part of the clearance is receptor-mediated.
  • Receptor-mediated endocytosis was studied using the Alexa Fluor 488 quenching techniques according to manufacturer's protocol (Life Technologies, Carlsbad, CA). Briefly, HEK cells expressing PD-L1 (HEK/PD-L1 cells) were incubated with 10 ⁇ g/ml Alexa Fluor 488-conjugated anti-PD-Ll /TGFp Trap on ice for about 1 hr and washed 4 times with cold media. Washed cells were then pulsed at 37 °C for 0.25, 0.5, 0.75, 1, 1.5, 2, 3 and 4 hr to allow internalization. Cell samples at each time point were then divided into two portions.
  • the receptor-mediated internalization kinetics is very similar to that of the anti-PD-Ll antibody, which is shown in FIG. 6B.
  • EXAMPLE 7 Anti-PD-Ll/TGFp Trap demonstrated a superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the EMT-6 (breast carcinoma) subcutaneous model [00171] 8-12 week old female Jh (Igh-T- 111101 TM) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.5 x 10 6 viable EMT6 cells in 0.1 ml PBS on the right flanks
  • Group 1 received 400 ⁇ g of isotype antibody control three times weekly (or "eod" (every other day);
  • Group 2 received 400 ⁇ g of anti-PD-Ll antibody three times weekly;
  • Group 3 received 164 ⁇ g of anti-PD- Ll(mut)/TGFp Trap three times weekly;
  • Group 4 received 492 ⁇ of anti-PD-Ll /TGFp Trap three times weekly;
  • Group 5 received 492 ⁇ g of anti-PD-L l /TGF Trap twice weekly
  • T/C ratio T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • FIG. 7A which showed the average tumor volumes of the surviving mice
  • FIG. 7B which showed the individual tumor volume of the surviving mice, noting that mice with tumors over 2500 mm 3 had to be euthanized.
  • Anti-PD-Ll /TGF Trap demonstrated potent anti-tumor efficacy, achieving T/C ratios of 0.30, 0.40, and 0.44 for the high (492 ⁇ g, Group 4), medium (164 ⁇ g, Group 6), and low (55 ⁇ g, Group 7) dose groups, respectively on Day 28.).
  • mice in a control group developed tumors to an average tumor volume of 726 mm 3 by Day 18 post challenge, none of the eleven mice previously treated with PD-Ll/TGFp Trap (three from Group 4, six from Group 5, and one each from Groups 6 and 7) showed any sign of tumor growth.
  • EXAMPLE 8 Anti-PD-Ll/TGF- ⁇ Trap showed profound synergistic anti-tumor activity in the MC38 (colorectal carcinoma) subcutaneous tumor model.
  • Group 1 received 400 ⁇ g of isotype antibody control;
  • Group 2 received 400 g of anti-PD-Ll antibody;
  • Group 3 received 133 ⁇ g of anti-PD-Ll antibody;
  • Group 4 received 492 ⁇ g of anti-PD-Ll (mut)/TGFp Trap;
  • Group 5 received 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap;
  • Group 6 received 492 ⁇ g of anti-PD- Ll /TGFp Trap;
  • Group 7 received 164 ⁇ g of anti-PD-Ll /TGF Trap.
  • the treatment was administered three times weekly for two weeks. Body weights were measured twice weekly to monitor toxicity.
  • EXAMPLE 9 Anti-PDLl/TGFp Trap was effective in the EMT-6 orthotopic model of metastatic breast cancer.
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 133 ⁇ g of anti-PD-Ll antibody;
  • Group 3 received 164 ⁇ g of anti-PD- Ll (mut)/TGFp Trap;
  • Group 4 received 164 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 5 received a combination of 133 ⁇ g of anti-PD-Ll and 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap.
  • Treatment was repeated on Days 0, 2, 4, 7, 9, 11 (i.e. 3 times weekly for two weeks). Body weights were measured twice weekly to monitor toxicity.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • the combination therapy of equimolar doses of anti-PD-Ll and anti-PD- Ll (mut)/TGFp Trap achieved almost identical anti-tumor efficacy as the fusion protein, although the exposure of the TGF Trap of the fusion protein (Group 4) was estimated to be about 3-fold lower than that of the anti-PD-Ll (mut)/TGFp Trap in the combination (Group 5) based on pharmacokinetics analysis (see Example 5).
  • tumors in Groups 4 and 5 continued to regress after the last day of dosing, e.g., average tumor size decreased from 212 mm 3 on Day 1 1, the last day of dosing, to 26 mm 3 on Day 24 for anti-PD- LI /TGF Trap treatment, demonstrating the long-lasting immunologic anti-tumor effect of targeting the two immunosuppressive mechanisms simultaneously.
  • EXAMPLE 10 Anti-PD-Ll/TGFp Trap has better anti-tumor efficacy than the combination of anti-PD-Ll and TGFp Trap in an intramuscular MC38 colorectal carcinoma model.
  • Group 1 received 400 ⁇ g of isotype antibody control;
  • Group 2 received 400 ⁇ g of anti- PD-Ll antibody;
  • Group 3 received 133 ⁇ g of anti-PD-Ll antibody;
  • Group 4 received 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap;
  • Group 5 received 492 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 6 received 164 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 7 received a combination of 133 ⁇ g of anti-PD-Ll and 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap.
  • Body weights were measured twice weekly to monitor toxicity.
  • Anti-PD-Ll (mut)/TGFp Trap at 164 ⁇ g (Group 4) was completely ineffective, and it should be pointed out that although this dose is equimolar with the low dose anti-PD- Ll/TGFp Trap group (Group 6), the exposure of the TGFP Trap should be fairly similar to that of the high dose anti-PD-Ll/TGFp Trap group (Group 5) because of the differences in pharmacokinetics (see Example 5). Therefore, the data demonstrated that anti-PD-Ll/TGFp Trap had potent synergistic anti-tumor activity in this model.
  • anti-PD-Ll/TGFp Trap was more efficacious than the combination therapy of equimolar doses of anti-PD-Ll and anti-PD-Ll (mut)/TGFp Trap, which had a T/C ratio of 0.16 (pO.001 vs. Group 1 and p>0.05 vs. Group 6) despite a higher TGFp Trap exposure of about threefold (see Example 5).
  • anti-PD-Ll /TGFp Trap treatment resulted in 4 out of 10 mice with complete tumor regression, while the combination of anti-PD-Ll and the Trap control induced complete regression in only 2 out of 10 mice (data not shown).
  • the tumors in the mice treated with anti-PD-Ll /TGFp Trap continued to regress after the last day of dosing on day 2, and stayed completely regressed thereafter (until at least Day 102), demonstrating the profound and long-lasting immunologic anti-tumor effect of this fusion protein.
  • the data supports a mechanism in which the anti-PD- LI /TGFP Trap fusion protein not only exploits the synergistic effect of blocking the two major immune escape pathways, but is superior to the combination therapy due to the targeting of the tumor microenvironment by a single molecular entity.
  • Many immunosuppressive cytokines secreted by tumor cells or subverted immune cells e.g.
  • anti-PD- L 1/TGFp Trap has the capability to deliver the TGFp Trap to the tumor microenvironment via binding to PD-L1+ tumor cells, where the Trap neutralizes the locally secreted TGFp.
  • anti- PD-Ll/TGFp Trap bound TGFp could be effectively destroyed through the PD-L1 receptor- mediated endocytosis (Examples 5 and 6).
  • EXAMPLE 11 Treatment with anti-PDLl/TGFp Trap or the combination of anti-PD- Ll and TGFp Trap control at equivalent exposure in the EMT-6 orthotopic model of metastatic breast cancer.
  • anti-PDLl/TGFp Trap had similar efficacy as the combination of anti-PD-Ll and TGFp Trap control in the orthotopic EMT-6 breast cancer model (Example 9).
  • efficacy of anti-PDLl/TGFp Trap or the combination of anti-PD- Ll and TGFp Trap control administered for equivalent exposure was tested.
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 164 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 3 received 55 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 4 received a combination of 133 of anti- PD-L1 and 55 ⁇ g of anti-PD-Ll (mut)/TGFp Trap;
  • Group 5 received a combination of 44.3 ⁇ g of anti-PD-Ll and 18.3 ⁇ g of anti-PD-Ll (mut)/TGFp Trap.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • EXAMPLE 12 Anti-PD-Ll/TGF- ⁇ Trap has better antitumor efficacy than the combination of anti-PD-Ll and TGFP Trap administered to give equivalent exposure in an intramuscular MC38 colorectal carcinoma model.
  • Example 10 The results in Example 10 suggested that at equimolar doses the anti-PD-Ll/TGF- ⁇ Trap has better antitumor efficacy than the combination of anti-PD-Ll and TGF Trap control even though the in vivo exposure of anti-PD-Ll (mut)/TGF Trap control is about 3 times that of anti-PD-Ll /TGFP Trap (Example 5).
  • Example 5 the anti-tumor efficacy of anti- PD-Ll/TGF Trap and the combination of anti-PD-Ll and anti-PD-Ll (mut)/TGFp Trap based on equal exposure was compared. Lower doses than in Example 10 were administered to avoid dosing near saturating levels.
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 164 ⁇ g of anti-PD-Ll/TGF Trap;
  • Group 3 received 55 ⁇ g of anti-PD-Ll /TGFp Trap;
  • Group 4 received a combination of 133 ⁇ g of anti- PD-Ll and 55 ⁇ g of anti-PD-Ll (mut)/TGFP Trap; and
  • Group 5 received a combination of 44.3 ⁇ g of anti-PD-Ll and 1 .3 ⁇ g of anti-PD-Ll (mut)/TGFp Trap.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • Anti-PD-Ll/TGF Trap demonstrated very potent anti-tumor efficacy, achieving T/C ratios of 0.13 (pO.001) and 0.19 (pO.001) for the intermediate (164 ⁇ g, Group 2, called intermediate dose relative to the high dose of 492 ⁇ g that seemed to be saturating in Example 10) and low (55 ⁇ g, Group 3) dose groups, respectively, on Day 9.
  • anti-PD-Ll /TGFP Trap was significantly more efficacious than the combination therapy of anti-PD-Ll and anti-PD-Ll (muf)/TGFp Trap at both dose levels (at the intermediate dose, T/C of 0.13 for anti-PD-Ll /TGFp Trap vs. 0.34 for the combination pO.0001 (Fig. 12B); at the low dose, T/C of 0.19 for anti-PD-Ll /TGFp Trap vs. 0.37 for the combination p ⁇ 0.0001 (Fig. 12C)).
  • EXAMPLE 13 Anti-PD-Ll(YW)/TGFp Trap has superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the EMT-6 (breast carcinoma) orthotopic model.
  • YW243.55S70 is a human antibody that recognizes both human and murine PD-Ll (US Patent Application Publication No. US2010/0203056 Al).
  • variable region sequence of the heavy chain (VH) and variable region sequence of the light chain (VL) were used to replace the corresponding variable region sequences of the anti-PD-Ll /TGFp Trap described in Example 1 to give anti-PD- Ll(YW)/TGFp Trap by standard molecular biology techniques.
  • the antibody fusion protein was expressed as described in Example 1.
  • the anti-PD-Ll antibody YW243.55S70 is similarly expressed for comparison of efficacy in murine tumor models.
  • Group 1 received 133 ⁇ of isotype antibody control;
  • Group 2 received 133 g of anti-PD-Ll(YW) antibody;
  • Group 3 received 164 ⁇ g of anti-PD-Ll(mut)/TGF Trap;
  • Group 4 received 164 ⁇ g of anti-PD-Ll(YW)/TGFp Trap;
  • Group 5 received a combination of 133 ⁇ g of anti-PD-Ll(YW) and 164 ⁇ g of anti-PD- LI (mut)/TGFp Trap.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • FIG. 13 A shows the average tumor volumes of the mice on Day 17, the last day for which the average tumor volume of all the mice were available, i.e., before mice with tumors over 2500 mm 3 were euthanized.
  • the anti-PD- Ll/TGFp Trap treated group had a median survival time of 65 days, which was significantly better than that of the anti-PD-Ll(YW) antibody treated group (24 days) or the TGF Trap control treated group (21 days). It also compares favorably with the median survival time of 53.5 days for the combination treatment group. Despite dosing stopped after day 7, the continual tumor growth inhibition and the prolonged survival of the anti-PD-Ll(YW)/TGFp Trap treated mice demonstrate the long-lasting immunologic anti-tumor effect resulting from dual blockade of the two major immunosuppressive pathways.
  • EXAMPLE 14 Anti-PD-Ll(YW)/TGF-p Trap has superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the MC38 (colorectal carcinoma) intramuscular tumor model
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 133 ⁇ g of anti- PD-Ll(YW) antibody;
  • Group 3 received 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap;
  • Group 4 received 164 ⁇ g of anti-PD-Ll(YW)/TGFp Trap;
  • Group 5 received a combination of 133 ⁇ g of anti-PD-Ll(YW) and 164 ⁇ g of anti-PD-L l(mut)/TGFp Trap.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • CT-01 1 also known as pidiluzumab, is a humanized anti-human PD1 antibody that was tested in the clinic for treatment of hematological malignancies (Berger et al, Clin Cancer Res. 2008; 14:3044-3051). It also recognizes murine PD-1 and has shown anti-tumor activity that synergizes with cyclophosphamide and vaccine treatment in syngeneic tumor models (Mkrtichyan et al., Eur J Immunol. 201 1; 41 :2977-86). The VH and VL sequences of CT-011 were used to produce a recombinant antibody with human IgGl /kappa constant regions by standard molecular biology techniques.
  • Group 1 received 364 ⁇ g of isotype antibody control;
  • Group 2 received 164 ⁇ g of anti-PD-Ll(mut)/TGFP Trap, which served as the TGF Trap control;
  • Group 3 received 200 ⁇ g of anti-PD-1 (CT-011);
  • Group 4 received a combination of 200 ⁇ g of anti-PD-1 (CT-01 1) and 164 g of anti-PD-Ll(mut)/TGFp Trap control.
  • T/C ratio the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • Group 1 received 364 ⁇ g of isotype antibody control on Days 0, 2, 4, and 7;
  • Group 2 received 164 ⁇ g of the anti-PD-Ll(mut)/TGFp Trap control on Days 0 and 2;
  • Group 3 received 200 ⁇ g of anti-PD-1 (CT-011) on Days 0, 2, 4, and 7;
  • Group 4 received a combination of 200 ⁇ g of anti-PD-1 (CT-01 1) on Days 0, 2, 4, and 7, and 164 ⁇ g of anti-PD-Ll(mut)/TGFp Trap control on Days 0 and 2.
  • T/C ratio T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • the anti-LAG3 antibody used is a rat IgGl monoclonal anti-murine LAG3 antibody C9B7W (BioXcell, Beverly, MA), which was shown to synergize with anti-murine PD-1 treatment in syngeneic tumor models (Woo et al, Cancer Res, 2011; 72:917-27).
  • the anti- TIM-3 antibody used is a rat IgG2a monoclonal anti-murine TIM3 antibody RMT3-23
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 164 ⁇ g of the anti-PD-Ll(mut)/TGFP Trap control;
  • Group 3 received 200 ⁇ g of anti-LAG3;
  • Group 4 received 250 ⁇ g of anti-TIM3;
  • Group 5 received a combination of 200 ⁇ g of anti-LAG3 and 164 ⁇ g of anti-PD-Ll(mut)/TGFp Trap control;
  • Group 6 received a combination of 250 ⁇ g of anti-TIM3 and 164 ⁇ g of anti-PD-Ll (mut)/TGFp Trap control.
  • T/C ratio T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
  • Anti-PD-Ll(mut)/TGFp Trap control (Group 2) showed very modest efficacy in this EMT-6 model.
  • Anti-T1M3 (Group 4) as a single agent showed a similarly modest efficacy as the Trap control, and in combination therapy with the Trap control (Group 6) showed no additive effect.
  • Anti-LAG3 either as a single agent (Group 3) or in combination therapy with the Trap control (Group 5) did not show any efficacy.
  • EXAMPLE 18 Combination treatment of TGFp Trap with either anti-LAG3 or anti- TIM-3 do not provide any additive anti-tumor effect in an MC38 (colorectal carcinoma) intramuscular tumor model
  • Group 1 received 133 ⁇ g of isotype antibody control;
  • Group 2 received 164 ⁇ g of the anti-PD-Ll(mut)/TGFp Trap control;
  • Group 3 received 200 ⁇ g of anti-LAG3;
  • Group 4 received 250 ⁇ g of anti-TIM3;
  • Group 5 received a combination of 200 ⁇ g of anti-LAG3 and 164 g of anti-PD-Ll(mut)/TGFp Trap control;
  • Group 6 received a combination of 250 ⁇ g of anti-TIM3 and 164 ⁇ g of anti-PD-Ll(mut)/TGFp Trap control.
  • Treatment was repeated on Days 2, 4, 7, 9, 1 1, 15 and 18. Body weights were measured twice weekly to monitor toxicity.
  • DNA sequence from the translation initiation codon to the translation stop codon of the anti- PD-L1 lambda light chain is the signal peptide from urokinase plasminogen activator
  • the leader sequence preceding the VL is the signal peptide from urokinase plasminogen activator
  • atgagggccctgctggctagactgctgctgtgcgtgctggtcgtgtccgacagcaagggcCAG TCCGCCCTGACCCAGCCTGCCTCCGTGTCTGGCTCCCCTGGCCAGTCCATCACCATCAGCTGC ACCGGCGACGTGGGCGGCTACAACTACGTGTCCTGGTATCAGCAGCACCCCGGC AAGGCCCCCAAGCTGATGATCTACGACGTGTCCAACCGGCCCTCCGGCGTGTCCAACAGATTC TCCGGCTCCAAGTCCGGCAACACCGCCTCCCTGACCATCAGCGGACTGCAGGCAGAGGACG

Abstract

D'une manière générale, la présente invention concerne des molécules bifonctionnelles comportant (a) le récepteur de type II de TGFβ ou un fragment de celui-ci capable de se lier à TGFβ et (b) un anticorps, ou fragment de liaison à l'antigène de celui-ci, qui se lie à une protéine de point de contrôle immunitaire, tel que le ligand de mort programmée 1 (PD-L1), les utilisations de ces molécules (par exemple, pour traiter le cancer), ainsi que des procédés de fabrication de telles molécules.
PCT/EP2015/052781 2014-02-10 2015-02-10 Inhibition ciblee de facteur de croissance de transformation bêta (tgfβ) WO2015118175A2 (fr)

Priority Applications (26)

Application Number Priority Date Filing Date Title
PE2020002093A PE20210168A1 (es) 2014-02-10 2015-02-10 INHIBICION DIRIGIDA DEL FACTOR DE CRECIMIENTO TRANSFORMADOR ß (TGFß)
BR112016014952A BR112016014952A2 (pt) 2014-02-10 2015-02-10 Inibição direcionada de tgfbeta
NZ721364A NZ721364A (en) 2014-02-10 2015-02-10 Targeted tgfβ inhibition
SG11201606577YA SG11201606577YA (en) 2014-02-10 2015-02-10 TARGETED TGFβ INHIBITION
EP15717097.8A EP3105246B1 (fr) 2014-02-10 2015-02-10 Inhibition ciblee de facteur de croissance de transformation bêta (tgfbeta)
DK15717097.8T DK3105246T3 (da) 2014-02-10 2015-02-10 Målrettet TGF-beta-inhibering
RS20210783A RS62038B1 (sr) 2014-02-10 2015-02-10 Ciljana tgf beta inhibicija
SI201531639T SI3105246T1 (sl) 2014-02-10 2015-02-10 Ciljana inhibicija TGF BETA
KR1020167024679A KR102363008B1 (ko) 2014-02-10 2015-02-10 표적화된 TGFβ 억제
CA2934979A CA2934979A1 (fr) 2014-02-10 2015-02-10 Inhibition ciblee de facteur de croissance de transformation beta (tgf?)
LTEP15717097.8T LT3105246T (lt) 2014-02-10 2015-02-10 Tikslinis tgf beta slopinimas
ES15717097T ES2876430T3 (es) 2014-02-10 2015-02-10 Inhibición dirigida de TGFß
CN201580007865.3A CN106103488B (zh) 2014-02-10 2015-02-10 靶向TGFβ抑制
AU2015213988A AU2015213988B2 (en) 2014-02-10 2015-02-10 Targeted TGFbeta inhibition
EP21165709.3A EP3889172A1 (fr) 2014-02-10 2015-02-10 Inhibition ciblee de facteur de croissance de transformation bêta (tgfbeta)
PL15717097T PL3105246T3 (pl) 2014-02-10 2015-02-10 Ukierunkowane hamowanie tgf beta
RU2016135062A RU2752424C2 (ru) 2014-02-10 2015-02-10 Направленное ингибирование tgf бета
JP2016546949A JP6731346B2 (ja) 2014-02-10 2015-02-10 標的TGFβ阻害
MX2016010067A MX2016010067A (es) 2014-02-10 2015-02-10 INHIBICION DIRIGIDA DEL FACTOR DE CRECIMIENTO TRANSFORMADOR ß (TGF ß).
IL246968A IL246968B (en) 2014-02-10 2016-07-27 Targeted inhibition of tgf–beta
PH12016501549A PH12016501549A1 (en) 2014-02-10 2016-08-04 Tergeted tgf� inhibition
AU2019246876A AU2019246876B2 (en) 2014-02-10 2019-10-11 Targeted TGFß Inhibition
IL281565A IL281565B (en) 2014-02-10 2021-03-16 Targeted inhibition of tgf-beta
HRP20210977TT HRP20210977T1 (hr) 2014-02-10 2021-06-18 Ciljano inhibiranje tgf beta
CY20211100564T CY1124408T1 (el) 2014-02-10 2021-06-23 ΣΤΟΧΕΥΜΕΝΗ ΑΝΑΣΤΟΛΗ TGFβ
AU2022200365A AU2022200365A1 (en) 2014-02-10 2022-01-20 Targeted TGFß Inhibition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461938048P 2014-02-10 2014-02-10
US61/938,048 2014-02-10

Publications (2)

Publication Number Publication Date
WO2015118175A2 true WO2015118175A2 (fr) 2015-08-13
WO2015118175A3 WO2015118175A3 (fr) 2015-10-08

Family

ID=52988007

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/052781 WO2015118175A2 (fr) 2014-02-10 2015-02-10 Inhibition ciblee de facteur de croissance de transformation bêta (tgfβ)

Country Status (27)

Country Link
US (4) US9676863B2 (fr)
EP (2) EP3889172A1 (fr)
JP (2) JP6731346B2 (fr)
KR (1) KR102363008B1 (fr)
CN (2) CN106103488B (fr)
AU (3) AU2015213988B2 (fr)
BR (1) BR112016014952A2 (fr)
CA (1) CA2934979A1 (fr)
CL (1) CL2016002017A1 (fr)
CY (1) CY1124408T1 (fr)
DK (1) DK3105246T3 (fr)
ES (1) ES2876430T3 (fr)
HR (1) HRP20210977T1 (fr)
HU (1) HUE054873T2 (fr)
IL (2) IL246968B (fr)
LT (1) LT3105246T (fr)
MX (2) MX2016010067A (fr)
NZ (1) NZ721364A (fr)
PE (2) PE20161096A1 (fr)
PH (1) PH12016501549A1 (fr)
PL (1) PL3105246T3 (fr)
PT (1) PT3105246T (fr)
RS (1) RS62038B1 (fr)
RU (2) RU2752424C2 (fr)
SG (2) SG10202108879SA (fr)
SI (1) SI3105246T1 (fr)
WO (1) WO2015118175A2 (fr)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017201036A1 (fr) * 2016-05-17 2017-11-23 Genentech, Inc. Signatures génétiques stromales destinées au diagnostic et à l'utilisation en immunothérapie
WO2018029367A1 (fr) * 2016-08-12 2018-02-15 Merck Patent Gmbh Polythérapie contre le cancer
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
WO2018208720A1 (fr) * 2017-05-09 2018-11-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combinaison de blocage de pdl1 et de tgf-bêta chez des patients atteints de malignités hpv +
WO2018205985A1 (fr) 2017-05-12 2018-11-15 江苏恒瑞医药股份有限公司 PROTÉINE DE FUSION CONTENANT UN RÉCEPTEUR DE TGF-β ET UTILISATIONS MÉDICALES ASSOCIÉES
EP3352800A4 (fr) * 2015-09-24 2019-03-27 The University of North Carolina at Chapel Hill Méthodes et compositions pour réduire les métastases
WO2019113464A1 (fr) * 2017-12-08 2019-06-13 Elstar Therapeutics, Inc. Molécules multispécifiques et utilisations correspondantes
WO2019175243A1 (fr) 2018-03-14 2019-09-19 Merck Patent Gmbh Composés et leurs utilisations pour traiter des tumeurs chez un sujet
WO2019211489A1 (fr) 2018-05-04 2019-11-07 Merck Patent Gmbh INHIBITION COMBINÉE DE PD-1/PD-L1, DE TGFβ ET D'ADN-PK POUR LE TRAITEMENT DU CANCER
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
WO2019242505A1 (fr) 2018-06-17 2019-12-26 上海健信生物医药科技有限公司 Anticorps ciblant cldn18.2, anticorps bispécifique, adc et car, et leurs applications
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
WO2020093023A1 (fr) 2018-11-01 2020-05-07 Merck Patent Gmbh Anticorps anti-tim-3
WO2020093024A2 (fr) 2018-11-01 2020-05-07 Merck Patent Gmbh Procédés d'administration d'anticorps anti-tim -3
WO2020094122A1 (fr) 2018-11-09 2020-05-14 江苏恒瑞医药股份有限公司 COMPOSITION PHARMACEUTIQUE À BASE D'UNE PROTÉINE DE FUSION DU RÉCEPTEUR TGF-β ET SON UTILISATION
JP2020514290A (ja) * 2017-01-07 2020-05-21 メルク パテント ゲーエムベーハー 標的tgf−β阻害のための投薬計画及び投薬形態
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10851157B2 (en) 2019-07-01 2020-12-01 Gensun Biopharma, Inc. Antagonists targeting the TGF-β pathway
WO2021084124A1 (fr) 2019-11-01 2021-05-06 Ares Trading S.A. Inhibition combinée de pd-1, tgfp et atm associée à une radiothérapie pour le traitement du cancer
WO2021089704A1 (fr) 2019-11-05 2021-05-14 Merck Patent Gmbh Inhibition combinée de pd-1, tgfb et tigit pour le traitement du cancer
EP3838260A1 (fr) 2019-12-20 2021-06-23 Ares Trading S.A. Composition de protéine de fusion rii igg:tgf
WO2021123432A1 (fr) 2019-12-20 2021-06-24 Ares Trading S.A. COMPOSITION DE PROTÉINE DE FUSION IGG : TGFβRII
CN113271962A (zh) * 2018-08-22 2021-08-17 默克专利有限公司 利用靶向TGF-β抑制的三阴性乳腺癌的治疗
WO2021169986A1 (fr) * 2020-02-25 2021-09-02 Wuxi Biologics (Shanghai) Co. Ltd. Protéine de fusion bifonctionnelle et ses utilisations
WO2021185337A1 (fr) 2020-03-20 2021-09-23 荣昌生物制药(烟台)股份有限公司 Protéine de fusion bispécifique et son utilisation
WO2021209358A1 (fr) 2020-04-14 2021-10-21 Glaxosmithkline Intellectual Property Development Limited Traitement combiné pour le cancer à base d'un anticorps icos et d'une protéine de fusion du récepteur tgf-bêta d'anticorps pd-l1
WO2021209458A1 (fr) 2020-04-14 2021-10-21 Ares Trading S.A. Traitement combiné du cancer
WO2022017487A1 (fr) 2020-07-24 2022-01-27 迈威(上海)生物科技股份有限公司 MUTANT DE RII DE TGF-β ET PROTÉINE DE FUSION ASSOCIÉE
WO2021263169A3 (fr) * 2020-06-26 2022-02-03 Sorrento Therapeutics, Inc. Virus oncolytiques exprimant des protéines hybrides immunomodulatrices
WO2022063114A1 (fr) * 2020-09-23 2022-03-31 海正生物制药有限公司 Mutant tgfbr2-ecd, protéine de fusion le comprenant et son utilisation
CN114349867A (zh) * 2020-10-14 2022-04-15 广东菲鹏制药股份有限公司 融合蛋白及其应用
EP3820496A4 (fr) * 2018-07-09 2022-04-20 Precigen, Inc. Constructions de fusion et leurs méthodes d'utilisation
WO2022090529A1 (fr) 2020-11-02 2022-05-05 Ares Trading S.A. Traitement combiné du cancer
WO2022090527A1 (fr) 2020-11-02 2022-05-05 Ares Trading S.A. Polythérapie du cancer
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11370819B2 (en) 2014-02-10 2022-06-28 Merck Patent Gmbh Targeted TGFβ inhibition
RU2776204C1 (ru) * 2017-05-12 2022-07-14 Цзянсу Хэнжуй Медицин Ко., Лтд. Слитый белок, содержащий рецептор tgf-бета, и его фармацевтическое применение
WO2022233718A2 (fr) 2021-05-03 2022-11-10 Merck Patent Gmbh Conjugués fragment-médicament de liaison à l'antigène fc ciblant her2
WO2022248380A1 (fr) 2021-05-25 2022-12-01 Merck Patent Gmbh Conjugués fragment de liaison à l'antigène fc-médicament ciblant l'egfr
WO2022258622A1 (fr) 2021-06-07 2022-12-15 Ares Trading S.A. Polythérapie anticancéreuse
US11583593B2 (en) 2016-01-14 2023-02-21 Synthis Therapeutics, Inc. Antibody-ALK5 inhibitor conjugates and their uses
EP4073124A4 (fr) * 2019-12-11 2024-01-24 Wuxi Biologics Ireland Ltd Anticorps bifonctionnel dirigé contre pd-l1 et tgfbeta
EP4107190A4 (fr) * 2020-02-18 2024-03-20 Nanjing Genscript Biotech Co Ltd Protéines de fusion et leurs utilisations

Families Citing this family (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150056199A1 (en) 2013-08-22 2015-02-26 Acceleron Pharma, Inc. Tgf-beta receptor type ii variants and uses thereof
MA42971A (fr) 2015-03-13 2018-08-15 Cytomx Therapeutics Inc Anticorps anti-pdl1, anticorps anti-pld1 activables, et leurs procédés d'utilisation
CN108136001B (zh) 2015-04-03 2022-07-29 佐马技术有限公司 使用TGF-β抑制剂和PD-1抑制剂治疗癌症
MA44594B1 (fr) 2015-05-29 2020-09-30 Memorial Sloan Kettering Cancer Center Anticorps anti-ctla-4 et méthodes d'utilisation de ceux-ci
AU2016301380B2 (en) 2015-08-04 2021-07-01 Acceleron Pharma Inc. Methods for treating myeloproliferative disorders
WO2017020291A1 (fr) * 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Nouveaux anticorps anti-pd-l1
EP3432927A4 (fr) 2016-03-24 2019-11-20 Gensun Biopharma Inc. Inhibiteurs trispécifiques pour le traitement du cancer
KR20230091191A (ko) 2016-05-27 2023-06-22 아게누스 인코포레이티드 항-tim-3 항체 및 이의 사용 방법
KR102617453B1 (ko) 2016-09-19 2023-12-26 현대모비스 주식회사 보조 표지판 인식 장치 및 그 방법
AU2017336546A1 (en) 2016-09-27 2019-05-02 Epicentrx, Inc. Immunomodulatory fusion proteins
WO2018071500A1 (fr) 2016-10-11 2018-04-19 Agenus Inc. Anticorps anti-lag-3 et leurs procédés d'utilisation
BR112019011651A2 (pt) 2016-12-07 2020-01-07 Agenus Inc. Anticorpos anti-cttla-4 e métodos de uso dos mesmos
CN110300599A (zh) 2016-12-07 2019-10-01 艾吉纳斯公司 抗体和其使用方法
WO2018128939A1 (fr) 2017-01-05 2018-07-12 Gensun Biopharma Inc. Antagonistes de régulateur de point de contrôle
KR20190102259A (ko) * 2017-01-10 2019-09-03 인트렉손 코포레이션 신규의 유전자 스위치 발현 시스템을 통한 폴리펩티드의 발현 조절
GB201700567D0 (en) 2017-01-12 2017-03-01 Genagon Therapeutics Ab Therapeutic agents
BR112019014510A2 (pt) 2017-01-13 2020-02-18 Agenus Inc. Receptores de célula t que se ligam ao ny-eso-1 e métodos de uso dos mesmos
AU2018228435A1 (en) 2017-03-02 2019-10-03 National Research Council Of Canada TGF-beta-receptor ectodomain fusion molecules and uses thereof
BR112019019939A2 (pt) * 2017-03-30 2020-04-28 Merck Patent Gmbh combinação de anticorpo anti-pd-l1 e um inibidor de dna-pk para tratamento de câncer
BR112019017241A2 (pt) 2017-04-13 2020-04-14 Agenus Inc anticorpos anti-cd137 e métodos de uso dos mesmos
US11021537B2 (en) 2017-05-01 2021-06-01 Agenus Inc. Anti-TIGIT antibodies and methods of use thereof
MD3628049T2 (ro) 2017-05-04 2023-10-31 Acceleron Pharma Inc Proteine de fuziune ale receptorului TGF-beta de tip II și utilizările acestora
WO2018222901A1 (fr) 2017-05-31 2018-12-06 Elstar Therapeutics, Inc. Molécules multispécifiques se liant à une protéine de leucémie myéloproliférative (mpl) et leurs utilisations
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof
JP7387585B2 (ja) 2017-09-04 2023-11-28 アジェナス インコーポレイテッド 混合系統白血病(mll)特異的ホスホペプチドに結合するt細胞受容体およびその使用方法
US11834492B2 (en) 2017-09-27 2023-12-05 Epicentrx, Inc. Human IL-10 receptor alpha fusion proteins
CN111213059B (zh) * 2017-11-06 2024-01-09 豪夫迈·罗氏有限公司 用于癌症的诊断和治疗方法
JP7319992B2 (ja) 2018-03-09 2023-08-02 アジェナス インコーポレイテッド 抗cd73抗体およびそれらの使用方法
WO2019178364A2 (fr) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Molécules multifonctionnelles et utilisations associées
WO2019178362A1 (fr) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Molécules multifonctionnelles se liant à calréticuline et utilisations associees
CA3096909A1 (fr) 2018-04-26 2019-10-31 Agenus Inc. Compositions peptidiques de liaison a une proteine de choc thermique (hsp) et leurs methodes d'utilisation
CA3099527A1 (fr) * 2018-05-03 2019-11-07 Acceleron Pharma Inc. Liants multispecifiques de ligands de la superfamille de tgfs et leurs utilisations
CA3099325A1 (fr) * 2018-05-03 2019-11-07 Acceleron Pharma Inc. Nouveaux liants de ligands de la superfamille des tgfs et leurs utilisations
AU2019271065A1 (en) * 2018-05-15 2020-11-05 Merck Patent Gmbh Dosing regimens for targeted TGF-B inhibition for use in treating cancer in treatment naive subjects
KR20210013096A (ko) 2018-05-17 2021-02-03 볼트 바이오테라퓨틱스 인코퍼레이티드 면역접합체
CA3103245A1 (fr) * 2018-06-13 2019-12-19 Merck Patent Gmbh Traitement du cancer bronchique non a petites cellules au stade iii et attenuation d'etats pathologiques associes au traitement
TW202019959A (zh) * 2018-06-22 2020-06-01 德商馬克專利公司 用於治療膽道癌的標靶性tgf-b抑制之給藥方案
JP2021529556A (ja) 2018-06-29 2021-11-04 ジェンサン バイオファーマ、 インコーポレイテッドGensun Biopharma, Inc. 抗腫瘍アンタゴニスト
AU2019299318A1 (en) * 2018-07-02 2021-01-21 Merck Patent Gmbh Combination therapy with targeted TGF-B inhibition for treatment of advanced non-small cell lung cancer
JP7465272B2 (ja) 2018-09-27 2024-04-10 マレンゴ・セラピューティクス,インコーポレーテッド Csf1r/ccr2多特異性抗体
SG11202105975SA (en) * 2018-12-06 2021-07-29 Guangdong Tcrcure Biopharma Technology Co Ltd Combinational tcr-t cell therapy targeting tumor antigens, tgf-beta, and immune checkpoints
AU2020226893A1 (en) 2019-02-21 2021-09-23 Marengo Therapeutics, Inc. Multifunctional molecules that bind to T cell related cancer cells and uses thereof
GB2597851A (en) 2019-02-21 2022-02-09 Marengo Therapeutics Inc Antibody molecules that bind to NKP30 and uses thereof
CN114127112A (zh) 2019-02-21 2022-03-01 马伦戈治疗公司 与t细胞结合的多功能分子及其治疗自身免疫性病症的用途
US20220226491A1 (en) 2019-03-15 2022-07-21 Bolt Biotherapeutics, Inc. Immunoconjugates Targeting PD-L1
AU2020290119A1 (en) * 2019-06-10 2021-10-07 Shandong Boan Biotechnology Co., Ltd. Bifunctional fusion protein against PDL1 and TGFβ and use thereof
CN114585390A (zh) 2019-06-13 2022-06-03 博尔特生物治疗药物有限公司 氨基苯并氮呯化合物、免疫缀合物及其用途
CN110218260A (zh) * 2019-06-24 2019-09-10 王跃驹 植物作为宿主在表达m7842中的应用
WO2021007428A2 (fr) * 2019-07-09 2021-01-14 The Johns Hopkins University Molécules, compositions et méthodes de traitement du cancer
CN114401995A (zh) * 2019-08-15 2022-04-26 南特比奥有限公司 TGF-β陷阱
EP4023679A4 (fr) 2019-08-30 2023-10-04 Nanjing Shunxin Pharmaceuticals Co., Ltd. of Chiatai Tianqing Pharmaceutical Group Protéine de fusion ciblant pd-l1 et tgf-beta et son utilisation
BR112022003740A2 (pt) 2019-08-30 2022-05-31 Agenus Inc Anticorpos anti-cd96 e métodos de uso dos mesmos
CN114630684A (zh) 2019-09-03 2022-06-14 博尔特生物治疗药物有限公司 氨基喹啉化合物、免疫缀合物及其用途
WO2021046347A1 (fr) 2019-09-04 2021-03-11 Bolt Biotherapeutics, Inc. Procédé de synthèse d'immunoconjugué
CN112442132A (zh) * 2019-09-05 2021-03-05 复旦大学 靶向肿瘤的重组双功能融合蛋白及其应用
MX2022003740A (es) 2019-09-30 2022-05-02 Bolt Biotherapeutics Inc Inmunoconjugados de aminobenzazepina unidos a amida y usos de estos.
KR20220071264A (ko) * 2019-09-30 2022-05-31 하버 바이오메드 (상하이) 컴퍼니 리미티드 항-pd-l1 항원 결합 단백질 및 이의 응용
CN114616251A (zh) * 2019-10-21 2022-06-10 南京维立志博生物科技有限公司 靶向PD-1和TGFβ的重组蛋白
AU2020369652A1 (en) 2019-10-25 2022-05-12 Bolt Biotherapeutics, Inc. Thienoazepine immunoconjugates, and uses thereof
WO2021093760A1 (fr) * 2019-11-12 2021-05-20 江苏恒瑞医药股份有限公司 PROTÉINE DE FUSION CONTENANT UN RÉCEPTEUR DE TGF-β, ET SON UTILISATION PHARMACEUTIQUE
WO2021104434A1 (fr) * 2019-11-27 2021-06-03 Shanghai Epimab Biotherapeutics Co., Ltd. PROTÉINES DE LIAISON BISPÉCIFIQUES DE TGFβ/PD-L1
CN113004414B (zh) * 2019-12-20 2022-09-06 广东菲鹏制药股份有限公司 抗PD1和TGFβ的双功能抗体及其制备方法,以及含有其的药物组合物
CN113121699A (zh) * 2019-12-30 2021-07-16 国典(北京)医药科技有限公司 Pttrap1双特异性融合蛋白及其应用
WO2021138407A2 (fr) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Molécules multifonctionnelles se liant à cd33 et utilisations associées
US20230086603A1 (en) 2020-01-21 2023-03-23 Bolt Biotherapeutics, Inc Anti-pd-l1 antibodies
EP4093772A1 (fr) 2020-01-21 2022-11-30 Bolt Biotherapeutics, Inc. Anticorps anti-pd-l1
KR20230028242A (ko) 2020-04-24 2023-02-28 마렝고 테라퓨틱스, 인크. T 세포 관련 암 세포에 결합하는 다중기능성 분자 및 그것의 용도
US11634477B2 (en) 2020-04-28 2023-04-25 The Rockefeller University Neutralizing anti-SARS-CoV-2 antibodies and methods of use thereof
BR112022020639A2 (pt) 2020-05-01 2022-11-29 Bolt Biotherapeutics Inc Anticorpos anti-dectina-2
US20230293716A1 (en) 2020-05-08 2023-09-21 Bolt Biotherapeutics, Inc. Elastase-substrate, peptide linker immunoconjugates, and uses thereof
US11028174B1 (en) 2020-07-28 2021-06-08 Lepu Biopharma Co., Ltd. Bifunctional molecules targeting PD-L1 and TGF-β
WO2022022503A1 (fr) * 2020-07-28 2022-02-03 Lepu Biopharma Co., Ltd. Molécules bifonctionnelles ciblant pd-l1 et tgf-bêta
WO2022042537A1 (fr) * 2020-08-24 2022-03-03 江苏恒瑞医药股份有限公司 APPLICATION D'UNE COMBINAISON D'UNE PROTÉINE DE FUSION DE RÉCEPTEUR DE TGF-β ET D'UN INHIBITEUR DE TYROSINE KINASE MULTICIBLE DANS LA PRÉPARATION D'UN MÉDICAMENT ANTITUMORAL
CA3190755A1 (fr) 2020-08-26 2022-03-03 Andreas Loew Molecules multifonctionnelles se liant a la calreticuline et utilisations associees
CN116761818A (zh) 2020-08-26 2023-09-15 马伦戈治疗公司 检测trbc1或trbc2的方法
KR20230074144A (ko) 2020-08-26 2023-05-26 마렝고 테라퓨틱스, 인크. NKp30에 결합하는 항체 분자 및 이의 용도
CN115884989A (zh) * 2020-08-31 2023-03-31 杭州九源基因工程有限公司 靶向PD-L1和TGFβ的双功能融合蛋白及其制备方法与应用
WO2022057061A1 (fr) * 2020-09-16 2022-03-24 Suzhou Neologics Bioscience Co., Ltd. Anticorps anti-pd-l1, protéines de fusion et leurs utilisations
WO2022057821A1 (fr) * 2020-09-16 2022-03-24 Suzhou Neologics Bioscience Co. , Ltd. Anticorps anti-pd-l1, protéines de fusion et leurs utilisations
WO2022063193A1 (fr) * 2020-09-24 2022-03-31 上海齐鲁制药研究中心有限公司 MOLÉCULE BIFONCTIONNELLE CIBLANT SIMULTANÉMENT PD-L1 ET TGFβ ET SON UTILISATION MÉDICALE
CN114573701A (zh) * 2020-12-02 2022-06-03 上海华奥泰生物药业股份有限公司 抗PD-L1/TGF-β双功能抗体及其用途
US20220195066A1 (en) 2020-12-11 2022-06-23 Bolt Biotherapeutics, Inc. Anti-cea immunoconjugates, and uses thereof
IL303491A (en) 2020-12-11 2023-08-01 Bolt Biotherapeutics Inc Anti-PD-L1 immunoconjugates and uses thereof
CN116635084A (zh) 2020-12-11 2023-08-22 博尔特生物治疗药物有限公司 抗cea免疫缀合物和其用途
WO2022174451A1 (fr) 2021-02-22 2022-08-25 浙江道尔生物科技有限公司 Protéine de fusion à domaines multiples ayant une activité anticancéreuse
CA3212345A1 (fr) 2021-03-02 2022-09-09 Glaxosmithkline Intellectual Property Development Limited Pyridines substituees utiles en tant qu'inhibiteurs de la dnmt1
EP4313162A1 (fr) 2021-03-26 2024-02-07 Bolt Biotherapeutics, Inc. Immunoconjugués de 2-amino-4-carboxamide-benzazépine et leurs utilisations
WO2022204528A1 (fr) 2021-03-26 2022-09-29 Bolt Biotherapeutics, Inc. Immunoconjugués de 2-amino-4-carboxamide-benzazépine et utilisations associées
AU2022335237A1 (en) * 2021-08-27 2024-03-28 Theranotics Co., Ltd. Bispecific molecule specifically binding to b7-h3 and tgfβ and uses thereof
US11945856B2 (en) 2022-01-28 2024-04-02 35Pharma Inc. Activin receptor type IIB variants and uses thereof
CN114990129B (zh) * 2022-05-11 2023-02-03 北京贝来生物科技有限公司 表达αPDL1:Fc融合蛋白的间充质干细胞的制备及应用

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058957A2 (fr) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Amelioration de la demi-vie circulante de proteines de fusion a base d'anticorps
US20030044423A1 (en) 2001-03-07 2003-03-06 Lexigen Pharmaceuticals Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
US20030166877A1 (en) 2001-03-30 2003-09-04 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5785682A (en) 1995-03-22 1998-07-28 Abbott Laboratories Pre-filled syringe drug delivery system
TR199902878T2 (xx) * 1997-04-18 2000-02-21 Biogen,Inc. Tip II TGF-Beta Resept�r/�m�noglob�lin sabit b�l�m f�zyon proteinleri.
JP2005112812A (ja) * 2003-10-09 2005-04-28 Zeria Pharmaceut Co Ltd リン酸化部位特異的抗体及びそれを用いたスクリーニング方法
KR20070007055A (ko) * 2003-12-24 2007-01-12 사이오스 인코퍼레이티드 Tgf-베타 억제제를 이용한 악성 신경교종의 치료법
US7670595B2 (en) 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
US7795389B2 (en) 2004-09-28 2010-09-14 The Board Of Regents Of The University Of Texas System Antagonizing TGF-beta activity with various ectodomains TGF-beta receptors used in combination or as fusion proteins
CN101541832B (zh) * 2006-09-07 2014-11-12 克鲁塞尔荷兰公司 能中和流感病毒h5n1的人结合分子及其应用
JP2010529859A (ja) 2007-06-15 2010-09-02 ジェンザイム、コーポレーション TGF−βII型受容体の2つのTGF−β結合ドメインを含有する融合タンパク質
US20140127200A1 (en) 2008-01-03 2014-05-08 The Scripps Research Institute Multispecific Antibody Targeting and Multivalency Through Modular Recognition Domains
WO2009152610A1 (fr) 2008-06-20 2009-12-23 The Royal Institution For The Advancement Of Learning/Mcgill University Conjugués d’interleukine-2 et de récepteur b de tgf-bêta de type ii soluble et leurs procédés et utilisations
CA2729810A1 (fr) 2008-07-02 2010-01-07 Emergent Product Development Seattle, Llc Proteines de liaison multi-cibles antagonistes du tgf-b
FR2944448B1 (fr) * 2008-12-23 2012-01-13 Adocia Composition pharmaceutique stable comprenant au moins un anticorps monodonal et au moins un polysacharide amphiphile comprenant des substituants derives d'alcools hydrofobes ou d'amines hydrophobes.
US9533055B2 (en) 2009-03-18 2017-01-03 Armagen Technologies, Inc. Compositions and methods for blood-brain barrier delivery of IgG-decoy receptor fusion proteins
EP2493929B1 (fr) 2009-10-28 2017-08-16 AbbVie Biotherapeutics Inc. Anticorps anti-egfr et leurs utilisations
KR101790767B1 (ko) 2009-11-24 2017-10-26 메디뮨 리미티드 B7―h1에 대한 표적화된 결합 물질
US20130129723A1 (en) * 2009-12-29 2013-05-23 Emergent Product Development Seattle, Llc Heterodimer Binding Proteins and Uses Thereof
CN103124743A (zh) * 2009-12-29 2013-05-29 新兴产品开发西雅图有限公司 Ron结合构建物及其使用方法
CN103237810A (zh) 2010-02-23 2013-08-07 霍夫曼-拉罗奇有限公司 用于治疗卵巢癌的抗血管发生疗法
DK2542590T4 (da) * 2010-03-05 2020-07-13 Univ Johns Hopkins Sammensætninger og fremgangsmåde til målrettede immunomodulatoriske antistof-fer og fusionproteiner
EA026924B1 (ru) * 2011-08-01 2017-05-31 Дженентек, Инк. Способы лечения рака с использованием антагонистов, связывающихся с осью pd-1, и ингибиторов mek
EP2844667B1 (fr) * 2012-04-30 2018-05-30 Biocon Limited Protéines de fusion ciblées/immunomodulatrices et leurs procédés de fabrication
BR112014029887A8 (pt) 2012-05-31 2021-09-14 Genentech Inc Método para tratar ou retardar a progressão do câncer, kits e uso de um antagonista de ligação do eixo pd-1, oxaliplatina, leucovorina e 5-fu
JP6720075B2 (ja) 2013-05-31 2020-07-08 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. 癌のための併用療法
CA2934028A1 (fr) 2013-12-17 2015-06-25 Genentech, Inc. Polytherapie comprenant des agonistes se liant a ox40 et des antagonistes se liant a l'axe pd-1
EP3889172A1 (fr) 2014-02-10 2021-10-06 Merck Patent GmbH Inhibition ciblee de facteur de croissance de transformation bêta (tgfbeta)
GB201403875D0 (en) 2014-03-05 2014-04-16 Cantargia Ab Novel antibodies and uses thereof
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
PT3180363T (pt) 2014-08-15 2019-11-05 Merck Patent Gmbh Proteínas de fusão de sirp-alfa e imunoglobulina

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058957A2 (fr) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Amelioration de la demi-vie circulante de proteines de fusion a base d'anticorps
US20030044423A1 (en) 2001-03-07 2003-03-06 Lexigen Pharmaceuticals Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
US20030166877A1 (en) 2001-03-30 2003-09-04 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
ANGAL S ET AL., MOL. IMMUNOL., vol. 30, 1993, pages 105 - 8
BARTRAM, CIRCULATION, vol. 103, 2001, pages 2745 - 52
BERGER ET AL., CLIN CANCER RES., vol. 14, 2008, pages 3044 - 3051
BIERIE ET AL., NAT REV CANCER, vol. 6, 2006, pages 506 - 20
BUJAK ET AL., CARDIOVASC RES., vol. 74, 2007, pages 184 - 95
CALONE ET AL., EXP ONCOL., vol. 34, 2012, pages 9 - 16
CONNOLLY ET AL., INT J BIOL SCI., vol. 8, 2012, pages 964 - 78
FLAVELL ET AL., NAT REV IMMUNOL., vol. 10, 2010, pages 554 - 67
GILLIES ET AL., CLIN CANCER RES., vol. 8, 2002, pages 210 - 6
HUME, BLOOD, vol. 119, 2012, pages 1810 - 20
KHAW ET AL., OPHTHALMOLOGY, vol. 114, 2007, pages 1822 - 1830
L6PEZ-CASILLAS ET AL., J CELL BIOL., vol. 124, 1994, pages 557 - 68
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LIN, J BIOL CHEM., vol. 270, 1995, pages 2747 - 54
MENDOZA ET AL., BIOCHEMISTRY, vol. 48, 2009, pages 11755 - 65
MKRTICHYAN ET AL., EUR J IMMUNOL., vol. 41, 2011, pages 2977 - 86
MONTERO-JULIAN ET AL., BLOOD, vol. 85, 1995, pages 917 - 24
MORRIS ET AL., J CLIN ONCOL, vol. 26, 2008, pages 9028
NGIOW ET AL., CANCER RES, vol. 71, 2011, pages 3540 - 51
PARDOLL, D.M., NAT IMMUNOL., vol. 13, 2012, pages 1129 - 32
PARDOLL, NAT REV CANCER, vol. 12, 2012, pages 253 - 264
ROBERTS ET AL., J CLIN INVEST., vol. 90, 1992, pages 2056 - 62
SUZUKI ET AL., CLIN CANCER RES., vol. 10, 2004, pages 5907 - 18
VERONA ET AL., PROTEIN ENG DES SEL., vol. 21, 2008, pages 463 - 73
VILCHIS-LANDEROS ET AL., BIOCHEM J, vol. 355, 2001, pages 215
WHERRY, E.J., NAT IMMUNOL., vol. 12, 2011, pages 492 - 9
WON ET AL., CANCER RES., vol. 59, 1999, pages 1273 - 7
WOO ET AL., CANCER RES, vol. 72, 2011, pages 917 - 27
YAMAGISHI, ANAT REC., vol. 295, 2012, pages 257 - 67
YANG ET AL., CANCER RES., vol. 68, 2008, pages 9107 - 11
YANG ET AL., TRENDS IMMUNOL, vol. 31, 2010, pages 220 - 227
YANG ET AL., TRENDS IMMUNOL., vol. 31, 2010, pages 220 - 227
ZHONG ET AL., CLIN CANCER RES., vol. 16, 2010, pages 1191 - 205

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11370819B2 (en) 2014-02-10 2022-06-28 Merck Patent Gmbh Targeted TGFβ inhibition
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
US10851165B2 (en) 2014-10-14 2020-12-01 Novartis Ag Antibody molecules to PD-L1 and methods of treating cancer
US11219694B2 (en) 2015-09-24 2022-01-11 The University Of North Carolina At Chapel Hill Methods and compositions for reducing metastases
EP3352800A4 (fr) * 2015-09-24 2019-03-27 The University of North Carolina at Chapel Hill Méthodes et compositions pour réduire les métastases
US11583593B2 (en) 2016-01-14 2023-02-21 Synthis Therapeutics, Inc. Antibody-ALK5 inhibitor conjugates and their uses
CN109312408B (zh) * 2016-05-17 2022-12-23 豪夫迈·罗氏有限公司 用于诊断和供免疫疗法中使用的基质基因签名
WO2017201036A1 (fr) * 2016-05-17 2017-11-23 Genentech, Inc. Signatures génétiques stromales destinées au diagnostic et à l'utilisation en immunothérapie
JP2019523218A (ja) * 2016-05-17 2019-08-22 ジェネンテック, インコーポレイテッド 免疫療法における診断及び使用のための間質遺伝子シグネチャー
JP7359547B2 (ja) 2016-05-17 2023-10-11 ジェネンテック, インコーポレイテッド 免疫療法における診断及び使用のための間質遺伝子シグネチャー
CN109312408A (zh) * 2016-05-17 2019-02-05 豪夫迈·罗氏有限公司 用于诊断和供免疫疗法中使用的基质基因签名
JP2019527706A (ja) * 2016-08-12 2019-10-03 メルク パテント ゲーエムベーハー 癌の併用療法
CN109641963A (zh) * 2016-08-12 2019-04-16 默克专利有限公司 癌症的联合治疗
WO2018029367A1 (fr) * 2016-08-12 2018-02-15 Merck Patent Gmbh Polythérapie contre le cancer
JP2020514290A (ja) * 2017-01-07 2020-05-21 メルク パテント ゲーエムベーハー 標的tgf−β阻害のための投薬計画及び投薬形態
US11440963B2 (en) 2017-05-09 2022-09-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination PDL1 and TGF-beta blockade in patients with HPV+ malignancies
WO2018208720A1 (fr) * 2017-05-09 2018-11-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combinaison de blocage de pdl1 et de tgf-bêta chez des patients atteints de malignités hpv +
US11274142B2 (en) 2017-05-12 2022-03-15 Jiangsu Hengrui Medicine Co., Ltd. Fusion protein containing TGF-β receptor and medicinal uses thereof
EP3623389A4 (fr) * 2017-05-12 2021-01-20 Jiangsu Hengrui Medicine Co., Ltd. Protéine de fusion contenant un récepteur de tgf- et utilisations médicales associées
WO2018205985A1 (fr) 2017-05-12 2018-11-15 江苏恒瑞医药股份有限公司 PROTÉINE DE FUSION CONTENANT UN RÉCEPTEUR DE TGF-β ET UTILISATIONS MÉDICALES ASSOCIÉES
RU2776204C1 (ru) * 2017-05-12 2022-07-14 Цзянсу Хэнжуй Медицин Ко., Лтд. Слитый белок, содержащий рецептор tgf-бета, и его фармацевтическое применение
KR20200004801A (ko) * 2017-05-12 2020-01-14 지앙수 헨그루이 메디슨 컴퍼니 리미티드 TGF-β 수용체를 함유하는 융합 단백질 및 이의 약학적 용도
KR102629503B1 (ko) * 2017-05-12 2024-01-24 지앙수 헨그루이 파마슈티컬스 컴퍼니 리미티드 TGF-β 수용체를 함유하는 융합 단백질 및 이의 약학적 용도
WO2019113464A1 (fr) * 2017-12-08 2019-06-13 Elstar Therapeutics, Inc. Molécules multispécifiques et utilisations correspondantes
WO2019175243A1 (fr) 2018-03-14 2019-09-19 Merck Patent Gmbh Composés et leurs utilisations pour traiter des tumeurs chez un sujet
WO2019211489A1 (fr) 2018-05-04 2019-11-07 Merck Patent Gmbh INHIBITION COMBINÉE DE PD-1/PD-L1, DE TGFβ ET D'ADN-PK POUR LE TRAITEMENT DU CANCER
WO2019242505A1 (fr) 2018-06-17 2019-12-26 上海健信生物医药科技有限公司 Anticorps ciblant cldn18.2, anticorps bispécifique, adc et car, et leurs applications
US11912763B2 (en) 2018-06-17 2024-02-27 L & L Biopharma Co., Ltd. Antibody targeting CLDN18.2, bispecific antibody, ADC, and CAR, and applications thereof
US11535669B2 (en) 2018-07-09 2022-12-27 Precigen, Inc. Fusion constructs and methods of using thereof
EP3820496A4 (fr) * 2018-07-09 2022-04-20 Precigen, Inc. Constructions de fusion et leurs méthodes d'utilisation
EP3840776A4 (fr) * 2018-08-22 2022-09-28 Merck Patent GmbH Traitement du cancer du sein triple négatif avec inhibition ciblée du tgf-b
CN113271962A (zh) * 2018-08-22 2021-08-17 默克专利有限公司 利用靶向TGF-β抑制的三阴性乳腺癌的治疗
WO2020093023A1 (fr) 2018-11-01 2020-05-07 Merck Patent Gmbh Anticorps anti-tim-3
WO2020093024A2 (fr) 2018-11-01 2020-05-07 Merck Patent Gmbh Procédés d'administration d'anticorps anti-tim -3
WO2020093024A3 (fr) * 2018-11-01 2020-06-04 Merck Patent Gmbh Procédés d'administration d'anticorps anti-tim -3
EP3878461A4 (fr) * 2018-11-09 2022-08-17 Jiangsu Hengrui Medicine Co., Ltd. COMPOSITION PHARMACEUTIQUE À BASE D'UNE PROTÉINE DE FUSION DU RÉCEPTEUR TGF-ß ET SON UTILISATION
WO2020094122A1 (fr) 2018-11-09 2020-05-14 江苏恒瑞医药股份有限公司 COMPOSITION PHARMACEUTIQUE À BASE D'UNE PROTÉINE DE FUSION DU RÉCEPTEUR TGF-β ET SON UTILISATION
WO2020263312A1 (fr) * 2019-06-28 2020-12-30 Gensun Biopharma, Inc. ANTAGONISTE ANTITUMORAL COMPOSÉ D'UN DOMAINE EXTRACELLULAIRE TGFβ1 - RII MUTÉ ET D'UN ÉCHAFAUDAGE D'IMMUNOGLOBULINE
JP2022540571A (ja) * 2019-06-28 2022-09-16 ゲンスン バイオファーマ、インコーポレーテッド 変異したTGFβ1-RII細胞外ドメインおよび免疫グロブリン足場で構成される抗腫瘍アンタゴニスト
US10851157B2 (en) 2019-07-01 2020-12-01 Gensun Biopharma, Inc. Antagonists targeting the TGF-β pathway
WO2021084124A1 (fr) 2019-11-01 2021-05-06 Ares Trading S.A. Inhibition combinée de pd-1, tgfp et atm associée à une radiothérapie pour le traitement du cancer
WO2021089704A1 (fr) 2019-11-05 2021-05-14 Merck Patent Gmbh Inhibition combinée de pd-1, tgfb et tigit pour le traitement du cancer
EP4073124A4 (fr) * 2019-12-11 2024-01-24 Wuxi Biologics Ireland Ltd Anticorps bifonctionnel dirigé contre pd-l1 et tgfbeta
EP3838260A1 (fr) 2019-12-20 2021-06-23 Ares Trading S.A. Composition de protéine de fusion rii igg:tgf
WO2021123432A1 (fr) 2019-12-20 2021-06-24 Ares Trading S.A. COMPOSITION DE PROTÉINE DE FUSION IGG : TGFβRII
EP4107190A4 (fr) * 2020-02-18 2024-03-20 Nanjing Genscript Biotech Co Ltd Protéines de fusion et leurs utilisations
EP4093779A4 (fr) * 2020-02-25 2024-03-06 Wuxi Biologics Ireland Ltd Protéine de fusion bifonctionnelle et ses utilisations
WO2021169986A1 (fr) * 2020-02-25 2021-09-02 Wuxi Biologics (Shanghai) Co. Ltd. Protéine de fusion bifonctionnelle et ses utilisations
WO2021185337A1 (fr) 2020-03-20 2021-09-23 荣昌生物制药(烟台)股份有限公司 Protéine de fusion bispécifique et son utilisation
WO2021209458A1 (fr) 2020-04-14 2021-10-21 Ares Trading S.A. Traitement combiné du cancer
WO2021209358A1 (fr) 2020-04-14 2021-10-21 Glaxosmithkline Intellectual Property Development Limited Traitement combiné pour le cancer à base d'un anticorps icos et d'une protéine de fusion du récepteur tgf-bêta d'anticorps pd-l1
WO2021263169A3 (fr) * 2020-06-26 2022-02-03 Sorrento Therapeutics, Inc. Virus oncolytiques exprimant des protéines hybrides immunomodulatrices
WO2022017487A1 (fr) 2020-07-24 2022-01-27 迈威(上海)生物科技股份有限公司 MUTANT DE RII DE TGF-β ET PROTÉINE DE FUSION ASSOCIÉE
WO2022063114A1 (fr) * 2020-09-23 2022-03-31 海正生物制药有限公司 Mutant tgfbr2-ecd, protéine de fusion le comprenant et son utilisation
CN114349867A (zh) * 2020-10-14 2022-04-15 广东菲鹏制药股份有限公司 融合蛋白及其应用
WO2022090527A1 (fr) 2020-11-02 2022-05-05 Ares Trading S.A. Polythérapie du cancer
WO2022090529A1 (fr) 2020-11-02 2022-05-05 Ares Trading S.A. Traitement combiné du cancer
WO2022233718A2 (fr) 2021-05-03 2022-11-10 Merck Patent Gmbh Conjugués fragment-médicament de liaison à l'antigène fc ciblant her2
WO2022248380A1 (fr) 2021-05-25 2022-12-01 Merck Patent Gmbh Conjugués fragment de liaison à l'antigène fc-médicament ciblant l'egfr
WO2022258622A1 (fr) 2021-06-07 2022-12-15 Ares Trading S.A. Polythérapie anticancéreuse

Also Published As

Publication number Publication date
US20180002436A1 (en) 2018-01-04
AU2015213988B2 (en) 2019-07-11
US11370819B2 (en) 2022-06-28
IL281565A (en) 2021-05-31
IL246968A0 (en) 2016-09-29
CN106103488A (zh) 2016-11-09
CN106103488B (zh) 2021-07-30
LT3105246T (lt) 2021-08-25
HRP20210977T1 (hr) 2021-09-17
AU2015213988A1 (en) 2016-07-07
KR20160119197A (ko) 2016-10-12
RU2016135062A (ru) 2018-03-15
JP2017506217A (ja) 2017-03-02
BR112016014952A2 (pt) 2017-09-19
DK3105246T3 (da) 2021-06-14
US20150225483A1 (en) 2015-08-13
CL2016002017A1 (es) 2017-06-09
AU2022200365A1 (en) 2022-02-17
CY1124408T1 (el) 2022-07-22
SG10202108879SA (en) 2021-09-29
SI3105246T1 (sl) 2021-11-30
JP6731346B2 (ja) 2020-07-29
EP3105246B1 (fr) 2021-03-31
WO2015118175A3 (fr) 2015-10-08
MX2021003685A (es) 2021-06-04
RU2016135062A3 (fr) 2018-12-12
KR102363008B1 (ko) 2022-02-16
IL246968B (en) 2021-04-29
IL281565B (en) 2021-12-01
NZ721364A (en) 2023-04-28
CN113549159A (zh) 2021-10-26
PE20210168A1 (es) 2021-01-28
SG11201606577YA (en) 2016-09-29
ES2876430T3 (es) 2021-11-12
CA2934979A1 (fr) 2015-08-13
PE20161096A1 (es) 2016-10-22
PT3105246T (pt) 2021-06-28
RU2752424C2 (ru) 2021-07-28
US20200055949A1 (en) 2020-02-20
MX2016010067A (es) 2016-10-07
RS62038B1 (sr) 2021-07-30
PH12016501549A1 (en) 2016-10-03
JP2020174674A (ja) 2020-10-29
AU2019246876B2 (en) 2021-10-21
AU2019246876A1 (en) 2019-10-31
PL3105246T3 (pl) 2021-11-29
HUE054873T2 (hu) 2021-10-28
EP3889172A1 (fr) 2021-10-06
EP3105246A2 (fr) 2016-12-21
RU2021114500A (ru) 2021-06-07
US20230056881A1 (en) 2023-02-23
US9676863B2 (en) 2017-06-13

Similar Documents

Publication Publication Date Title
US11370819B2 (en) Targeted TGFβ inhibition
US20220017621A1 (en) Combination Therapy for Cancer
RU2750675C1 (ru) Антитела против pd-1 и композиции
US20190330375A1 (en) Dosing regimens and dosage forms for targeted tgf-b inhibition
KR102526945B1 (ko) Cd269에 대한 인간화 항체
IL293751A (en) Heavy chain antibodies that bind to cd38
US20240025967A1 (en) Recombinant protein targeting pd-1 and tgfbeta
KR20150122730A (ko) 암의 치료에서 항-gcc 항체-약물 콘주게이트 및 dna 손상 제제의 투여
CN114867751A (zh) 4-1bb和ox40结合蛋白及相关组合物和方法、抗-4-1bb抗体、抗-ox40抗体

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15717097

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2934979

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 122020025158

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2015717097

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015717097

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016014952

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2015213988

Country of ref document: AU

Date of ref document: 20150210

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016546949

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 001267-2016

Country of ref document: PE

WWE Wipo information: entry into national phase

Ref document number: 246968

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/010067

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 12016501549

Country of ref document: PH

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15717097

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20167024679

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201609356

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2016135062

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016014952

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160624